Skip to main content

Full text of "USPTO Patents Application 10551504"

See other formats


(19) 



J 



Europalsches 
Patentamt 



European 
Patent Office 



Office europeen 
des brevets 



(11) 



EP 1 757 686 A1 



(12) 



EUROPEAN PATENT APPLICATION 

published in accordance with Art. 158(3) EPC 



(43) Date of publication: 

28.02.2007 Bulletin 2007/09 

(21 ) Application number: 04726750.5 

(22) Date of filing: 09.04.2004 



(51) IntCI.: 

C12N 15/09 (2008.01) C(J7K 16/28 (2006.01) 

C07K 16/46 (2006.01) A61 p 35/00 (2006.01) 

A61P 37/02 (2006.01) A61P 43/00 (2006.01) 
A61K 39/395 f 2006 01 ) 

(86) International application number: 
PCT/JP2004/005152 



(87) International publication number: 

WO 2005/100560 (27.10.2005 Gazette 2005/43) 



(84) 


Designated Contracting States: 


• ABE, Masahiro 




AT BE BG CH CY CZ DE DK EE ES Fl FR GB GR 


Tokushima-shi, Tokushima 7700033 (JP) 




HU IE IT LI LU MC NL PL PT RO SE SI SK TR 


• TSUCHIYA, Masayuki, 






CHUGAI SEIYAKU KABUSHIKI K. 


(71) 


Applicants: 


Gotenba-shi, Shizuoka 4128513 (JP) 




CHUGAI SEIYAKU KABUSHIKI KAISHA 


• KIMURA, Naoki, 




Tokyo, 115-8543 (JP) 


CHUGAI SEIYAKU KABUSHIKI KAISHA 




Ozaki, Shuji 


Gotenba-shi, Shizuoka 4128513 3 (JP) 




Tokushima-shi, Tokushima 770-0804 (JP) 


• KAWAI, Shigeto, 




Abe, Masahiro 


CHUGAI SEIYAKU KABUSHIKI KAISHA 




Tokushima-shi, Tokushima 770-0033 (JP) 


Gotenba-shi, Shizuoka 4128513 (JP) 


(72) 


Inventors: 


(74) Representative: Vossius & Partner 




OZAKI, Shuji 


Siebertstrasse 4 




Tokushima-shi, Tokushima 7700804 (JP) 


81675 Munchen (DE) 



(54) CELL DEATH INDUCER 



(57) To identify antigens of the 2D7 antibody, the 
present inventors cloned the 2D7 antigen. As a result, 
the 2D7 antibody was found to recognize HLA class I A. 
In addition, the present inventors examined whether the 
2D7 antibody has cell death-inducing activity. Nuclei frag- 
mentation was observed when the 2D7 antibody was 



cross-linked with another antibody, indicating that cell- 
death was induced. Further, diabodies of the 2D7 anti- 
body were found to have very strong cell death-inducing 
activities, even without the addition of another antibody. 
These results indicate that minibodies of an HLA-recog- 
nizing antibody can be used as cell death-inducing 
agents. 



FIG. 10A 

LEADER SEQUENCE 



00 
CD 

LO 




Printed by Jouve, 75001 PARIS (FR) 



(Cont. next page) 



EP 1 757 686 A1 



FIG. 10B 




FIG. 10C LYSATES CULTURE 

LYbAitb syPERNATANTS 

CD ^ CQ 

I t*~- I h- 

uj Q uj O 
> CM > CM 



EP 1 757 686 A1 



Description 

Technical Field 

5 [0001] The present invention relates to minibodies that recognize HLA. 
Background Art 

[0002] The HLA class I antigen is formed by a heterodimer of a 45-KD a chain comprising three domains (cc1, oc2, 
10 oc3), and a 12-KD (32 microglobulin. The main role of the HLA molecule is to present CD8+T cells with antigenic peptides 
formed from about eight to ten amino acids and produced inside cells. As such, it plays a very important role in the 
immune response and immune tolerance induced by this peptide presentation. 

[0003] Cell growth-suppressing and cell death-inducing effects have been observed in lymphocytes upon HLA class 
IA antigen and antibody ligation, suggesting that HLA molecules may also be signal transduction molecules. 

15 [0004] More specifically, for example, there are reports showing cell growth suppression of activated lymphocytes by 
the B9.12.1 antibody against the oil domain of human HLA class IA, the W6/32 antibody against the oc2 domain, and 
the TP25.99 and A1.4 antibodies against the oc3 domain (Non-patent Documents 1, 2). Furthermore, two types of 
antibodies, MoAb90 and YTH862, against the human HLA class IA oc1 domain have been reported to induce apoptosis 
in activated lymphocytes (Non-patent Documents 2, 3, 4). Apoptosis induced by these two antibodies has been shown 

20 to be a caspase-mediated reaction (Non-patent Document 4), and therefore, HLA class IA antigens expressed in lym- 
phocytes are also speculated to be involved in apoptosis signal transduction. 

[0005] Furthermore, the 5H7 antibody against the oc3 domain of human HLA class IA (Non-patent Document 5), and 
the RE2 antibody against the a2 domain of mouse HLA class IA (Non-patent Document 6) have been also reported to 
induce cell death in activated lymphocytes and the like. However, in contrast with the aforementioned apoptosis-inducing 
25 antibodies MoAb90 and YTH862, it has been shown that none of the cell deaths induced by these antibodies are caspase- 
mediated. Accordingly, cell deaths due to 5H7 and RE2 are predicted to be of a type completely different from conven- 
tionally known apoptosis mechanisms. 

[0006] As described above, there are numerous reports of the cell growth-suppressing actions and cell death-inducing 
actions of anti-HLA antibodies. However, the antibodies used herein are all in the molecular forms of IgG antibodies, F 
so (ab')2, or Fab, and to date there have been no reports that cell death-inducing activity is enhanced by reducing the 
molecular weight of antibodies, as in F(ab')2 and Fab. 
[0007] Prior art literature relating to the present invention is shown below: 

[Non-patent Document 1] Fayen et al., Int. Immunol. 10: 1347-1358(1998) 
35 [Non-patent Document 2] Genestier et al., Blood 90: 3629-3639 (1 997) 

[Non-patent Document 3] Genestier et al., Blood 90: 726-735 (1 997) 

[Non-patent Document 4] Genestier et al., J. Biol. Chem. 273: 5060-5066 (1998) 

[Non-patent Document 5] Woodle et al., J. Immunol. 158: 2156-2164 (1997) 

[Non-patent Document 6] Matsuoka et al., J. Exp. Med. 181 : 2007-2015 (1995) 
40 [Non-patent Document 7] Goto, et al. Blood 84: 1 922 (1 994) 

Disclosure of the Invention 

[0008] The primary purpose of this invention is to provide minibodies of antibodies that recognize HLA class IA. A 
45 further objective of this invention is to provide novel therapeutic agents for tumors or autoimmune diseases that utilize 
these minibodies. 

[0009] The present inventors obtained 2D7 antibodies recognizing HLA class IA. Then, it was examined whether the 
2D7 antibodies have cell death-inducing activity. More specifically, Jurkat cells were cultured in the presence or absence 
of 2D7, with anti-mouse IgG antibody also added. Cell nuclei were stained 48 hours later with Hoechst 33258, and then 

so checked for cell nuclei fragmentation, which is characteristic of dead cells. As a result, hardly any cell death-inducing 
activity was observed in Jurkat cells with 2D7 antibody alone; however, nuclei fragmentation was observed when the 
antibody was further cross-linked with anti-mouse IgG antibody, showing that cell death was induced. 
[0010] As described, because cross-linking with an anti-mouse IgG antibody is necessary for 2D7 antibody to induce 
cell death, it is difficult to clinically apply the 2D7 antibody to tumors or autoimmune diseases. Therefore, the present 

55 inventors examined the effect of reducing the molecular weight of the 2D7 antibody on cell death induction. More 
specifically, genes encoding the variable regions of the 2D7 antibody were cloned from hybridomas. The 2D7 antibody 
was then made into diabodies using genetic engineering techniques and effects on cell death-inducing activity were 
examined. Surprisingly, the 2D7 antibody converted to diabodies showed strong cell death-inducing activity within a 



3 



EP 1 757 686 A1 



very short time and at low doses, even without cross-linking with an anti-mouse IgG antibody. Furthermore, the diabody 
hardly acted on normal peripheral blood-derived lymphocytes and adherent cells, and specifically induced cell death in 
various myeloma cells, T cell leukemia cell lines, and activated lymphocytes. The above-mentioned results show that 
the minibodies of antibodies recognizing HLA can be utilized as cell death-inducing agents. 
5 [0011] More specifically, the present invention provides the following [1] to [14]: 

[I] a minibody, which comprises a heavy chain variable region comprising CDRs 1, 2, and 3 which consist of the 
amino acid sequences of SEQ ID NOs: 13, 14, and 15; 

[2] a minibody which is functionally equivalent to the minibody of [1], and which comprises heavy chain CDRs 
10 consisting of amino acid sequences with one or more amino acid substitutions, deletions, insertions, and/or additions 

in the amino acid sequences of the heavy chain CDRs of the minibody of [1 ]; 

[3] a minibody, which comprises a light chain variable region comprising CDRs 1, 2, and 3 which consist of the 
amino acid sequences of SEQ ID NOs: 16, 17, and 1 8; 

[4] a minibody which is functionally equivalent to the minibody of [3], and which comprises light chain CDRs consisting 
15 of amino acid sequences with one or more amino acid substitutions, deletions, insertions, and/or additions in the 

amino acid sequences of the light chain CDRs of the minibody of [3]; 

[5] a minibody, which comprises a heavy chain variable region comprising CDRs 1, 2, and 3 which consist of the 
amino acid sequences of SEQ ID NOs: 13, 14, and 15, and a light chain variable region comprising CDRs 1,2, and 
3 which consist of the amino acid sequences of SEQ ID NOs: 16, 17, and 1 8; 
20 [6] a minibody which is functionally equivalent to the minibody of [5], and which comprises CDRs consisting of amino 

acid sequences with one or more amino acid substitutions, deletions, insertions, and/or additions in the amino acid 
sequences of the CDRs of the minibody of [5]; 
[7] the minibody of any one of [1 ] to [6], which is a diabody; 

[8] a cell death-inducing agent comprising the minibody of any one of [1] to [7] as an active ingredient; 
25 [9] the cell death-inducing agent of [8], which induces cell death of a B-cell or T-cell; 

[10] the cell death-inducing agent of [9], wherein the B-cell or T-cell is an activated B-cell or an activated T-cell; 

[I I] a cell growth-suppressing agent comprising the minibody of any one of [1 ] to [7] as an active ingredient; 
[12] an anti-tumor agent comprising the minibody of any one of [1] to [7] as an active ingredient; 

[13] the anti-tumor agent of [12], wherein the tumor is blood tumor; and 
30 [14] an agent for treating an autoimmune disease, wherein the agent comprises the minibody of any one of [1 ] to 

[7] as an active ingredient. 

[0012] The present invention provides minibodies that recognize HLA. The minibodies of this invention are useful 
since their activity is elevated. Herein activity refers to a biological action that is caused by binding an antibody to an 
35 antigen. Specific examples include cell death-inducing actions, apoptosis-inducing actions, cell growth-suppressing 
actions, cell differentiation-suppressing actions, cell division-suppressing actions, cell growth-inducing actions, cell dif- 
ferentiation-inducing actions, cell division-inducing actions, and cell cycle-regulating actions. Cell death-inducing actions 
and cell growth-suppressing actions are preferred. 

[0013] The cells that become the target of the above-mentioned actions, such as cell death-inducing actions and cell 
40 growth-suppressing actions, are not particularly limited, though blood cells and non-adherent cells are preferred. Specific 
examples of blood cells include lymphocytes (B cells, T cells), neutrophils, eosinophils, basophils, monocytes (preferably 
activated peripheral blood mononuclear cells (PBMC)), and myeloma cells, while lymphocytes (B cells, T cells), and 
myeloma cells are preferred, and T cells or B cells (particularly activated B cells or activated T cells) are most preferable. 
Non-adherent cells refer to cells that, when cultured, grow in a suspended state without adhering to the surface of 
45 culturing vessels of glass, plastic or the like. On the other hand, adherent cells refer to cells that, when cultured, adhere 
to the surface of culturing vessels of glass, plastic or the like. 

[0014] In the present invention, administration of the minibodies that recognize HLA can treat or prevent diseases 
such as tumors, including blood tumors (hematopoietic tumors) (specific examples include leukemia, myelodysplastic 
syndrome, malignant lymphoma, chronic myelogenic leukemia, plasmacytic disorders (myeloma, multiple myeloma, 

so macroglobulinemia), and myeloproliferative diseases (polycythemia vera, essential thrombocythemia, idiopathic mye- 
lofibrosis)), and autoimmune diseases (specific examples include rheumatism, autoimmune hepatitis, autoimmune thy- 
roiditis, autoimmune bullosis, autoimmune adrenocortical disease, autoimmune hemolytic anemia, autoimmune throm- 
bycytopenic purpura, autoimmune atrophic gastritis, autoimmune neutropenia, autoimmune orchitis, autoimmune en- 
cephalomyelitis, autoimmune receptor disease, autoimmune infertility, Crohn's disease, systemic lupus erythematosus, 

55 multiple sclerosis, Basedow's disease, juvenile diabetes, Addison's disease, myasthenia gravis, lens-induced uveitis, 
psoriasis, and Behchet's disease). 

[0015] In the present invention, HLA refers to human leukocyte antigen. HLA molecules are categorized into class I 
and class II. Known examples of class I are HLA-A, B, C, E, F, G, H, J, and such; and known examples of class II are 



4 



EP 1 757 686 A1 



HLA-DR, DQ, DP, and such. The antigens recognized by the antibodies of this invention are not particularly limited, so 
long as they are HLA molecules, preferably molecules classified as class I, and more preferably HLA-A. 
[0016] In the present invention, a minibody comprises an antibody fragment that lacks a portion of a whole antibody 
(for example, whole IgG). The minibodies of the present invention are not particularly limited so long as they can bind 

5 an antigen. There are no particular limitations on the antibody fragments of the present invention, so long as they are 
portions of a whole antibody, and preferably contain a heavy chain variable region (VH) or a light chain variable region 
(VL). More preferably, the antibody fragments contain both a heavy chain variable region (VH) and a light chain variable 
region (VL). Specific examples of the antibody fragments include Fab, Fab', F(ab')2, Fv, and scFv (single chain Fv), but 
are preferably scFv (Huston, J. S. et al., Proc. Natl. Acad. Sci. U.S.A. (1 988)85, 5879-5883; Plickthun "The Pharmacology 

io of Monoclonal Antibodies" Vol. 1 13, Resenburg and Moore Ed., Springer Verlag, New York, pp. 269-31 5, (1 994)). Such 
antibody fragments can be prepared by treating an antibody with an enzyme, such as papain or pepsin for example, to 
generate antibody fragments, or by constructing genes that encode these antibody fragments, introducing them into 
expression vectors, and then expressing them in appropriate host cells (see, for example, Co, M. S. et al., 1994, J. 
Immunol. 152, 2968-2976; Better, M. and Horwitz, A. H., 1989, Methods Enzymol. 178, 476-496; Pluckthun, A. and 

15 Skerra, A., 1989, Methods Enzymol. 178, 497-515; Lamoyi, E., 1986, Methods Enzymol. 121, 652-663; Rousseaux, J. 
et al., 1986, Methods Enzymol. 121, 663-669; Bird, R. E. and Walker, B. W., 1991, Trends Biotechnol. 9,132-137). 
[0017] The minibodies of this invention preferably have smaller molecular weights than a whole antibody; however, 
they may form multimers, including dimers, trimers, and tetramers, and the molecular weights may become greater than 
that of the whole antibody. 

20 [0018] A preferred minibody of this invention is an antibody comprising two or more antibody VHs and two or more 
antibody VLs, in which each of these variable regions is linked directly or indirectly via linkers and such. Such linkages 
may be covalent bonds or non-covalent bonds, or may be both. An even more preferable minibody is an antibody 
comprising two or more VH-VL pairs formed by non-covalent bonding between VH and VL. In this case, the distance 
between one VH-VL pair and another VH-VL pair is preferably shorter in a minibody than in a whole antibody. 

25 [0019] A particularly preferable minibody of this invention is a diabody. A diabody is a dimer formed by binding two 
fragments, in which a variable region is linked to another variable region via a linker and such (for example, scFv) 
(hereinafter referred to as diabody-constituting fragments), and usually comprises two VLs and two VHs (P. Holliger et 
al., Proc. Natl. Acad. Sci. USA, 90, 6444-6448 (1 993); EP404097; W093/1 1161; Johnson et al., Method in Enzymology, 
203, 88-98, (1991); Holliger et al., Protein Engineering, 9, 299-305, (1996); Perisic et al., Structure, 2, 1217-1226, (1994) 

so ■ John et al., Protein Engineering, 12(7), 597-604, (1999); Holliger et al., Proc. Natl. Acad. Sci. USA., 90, 6444-6448, 
(1 993); Atwell et al., Mol. Immunol. 33, 1301-1312, (1996)). The bonds between the diabody-constituting fragments may 
be non-covalent or covalent bonds, but are preferably non-covalent bonds. 

[0020] Alternatively, diabody-constituting fragments may be bound by a linker and such to form a single chain diabody 
(sc diabody). In such cases, linking the diabody-constituting fragments using a long linker of about 20 amino acids allows 

35 non-covalent bond formation between diabody-constituting fragments on the same chain, thus forming a dimer. 

[0021] Diabody-constituting fragments include those with a linked VL-VH, linked VL-VL, and linked VH-VH, and are 
preferably those with a linked VH-VL. In the diabody-constituting fragments, the linker used to link a variable region to 
a variable region is not particularly limited, but is preferably a linker short enough to prevent non-covalent bonding 
between variable regions in the same fragment. The length of such a linker can be appropriately determined by those 

40 skilled in the art, and is ordinarily 2 to 14 amino acids, preferably 3 to 9 amino acids, and most preferably 4 to 6 amino 
acids. In this case, linkers between a VL and VH encoded on the same fragment are short, and thus a VL and VH on 
the same strand do not form a non-covalent bond nor a single-chain V region fragment; rather, the fragment forms a 
dimer with another fragment via non-covalent bonding. Furthermore, according to the same principle as in diabody 
construction, three or more diabody-constituting fragments may be bound to form multimeric antibodies, such as trimers 

45 and tetramers. 

[0022] The present invention's minibodies recognizing HLA-A include minibodies that contain heavy chain variable 
regions comprising CDRs 1, 2, and 3 which consist of the amino acid sequences of SEQ ID NOs: 13, 14, and 15, 
respectively. Also included are minibodies that are functionally equivalent to the above minibodies and which comprise 
heavy chain CDRs consisting of amino acid sequences with one or more amino acid substitutions, deletions, insertions, 

so and/or additions in the heavy chain CDR amino acid sequences of the above minibodies. 

[0023] In addition, the minibodies of the present invention include minibodies that contain light chain variable regions 
comprising CDRs 1 , 2, and 3 which consist of the amino acid sequences of SEQ ID NOs: 16, 17, and 1 8, respectively. 
Also included are minibodies that are functionally equivalent to the above minibodies and which comprise light chain 
CDRs consisting of amino acid sequences with one or more amino acid substitutions, deletions, insertions, and/or 

55 additions in the light chain CDR amino acid sequences of the above minibodies. 

[0024] Preferred examples of the minibodies of the present invention include minibodies that contain heavy chain 
variable regions comprising CDRs 1 , 2, and 3 which consist of the amino acid sequences of SEQ ID NOs: 13, 14, and 
15, respectively, and light chain variable regions comprising CDRs 1 , 2, and 3 which consist of the amino acid sequences 



5 



EP 1 757 686 A1 



of SEQ ID NOs: 16, 17, 18. 

[0025] Furthermore, preferred examples of the minibodies include minibodies that are functionally equivalent to the 
above-described minibodies and which comprise CDRs consisting of amino acid sequences with one or more amino 
acid substitutions, deletions, insertions, and/or additions in the CDR amino acid sequences of the above-described 
5 minibodies. 

[0026] A particularly preferred minibody of the present invention include a diabody that comprises the following amino 
acid sequences: AspTyrPhelleHis (SEQ ID NO: 13) as heavy chain CDR1, TrpllePheProGlyAspAspThrThrAsp- 
TyrAsnGluLysPheArgGly(SEQ ID NO: 14) as heavy chain CDR2, SerAspAspPheAspTyr (SEQ ID NO: 15) as heavy 
chain CDR3, SerAlaSerSerSerValSerTyrMetHis (SEQ ID NO: 16) as light chain CDR1, SerThrSerAsnLeuAlaSer (SEQ 
10 ID NO: 1 7) as light chain CDR2, and GlnGlnArgThrSerTyrProProThr (SEQ ID NO: 1 8) as light chain CDR3. 

[0027] Herein, "functionally equivalent" means that the minibody of interest has an activity equivalent to that of a 
diabody of interest (for example, HLA-A binding activity, and cell death-inducing activity). 

[0028] The number of mutated amino acids is not particularly limited, but may usually be 30 amino acids or less, 
preferably 15 amino acids or less, and more preferably five amino acids or less (for example, three amino acids or less). 

15 The amino acids are preferably mutated or modified in a way that conserves the properties of the amino acid side chain. 
Examples of amino acid side chain properties are: hydrophobic amino acids (A, I, L, M, F, P, W, Y, and V), hydrophilic 
amino acids (R, D, N, C, E, Q, G, H, K, S, and T), amino acids comprising the following side chains: aliphatic side chains 
(G, A, V, L, I, and P); hydroxyl-containing side chains (S, T, and Y); sulfur-containing side chains (C and M); carboxylic 
acid- and amide-containing side chains (D, N, E, and Q); basic side chains (R, K, and H); aromatic ring-containing side 

20 chains (H, F, Y, and W) (amino acids are represented by one-letter codes in parentheses). Polypeptides comprising a 
modified amino acid sequence, in which one or more amino acid residues is deleted, added, and/or replaced with other 
amino acids, are known to retain their original biological activities (Mark, D. F. et al., Proc. Natl. Acad. Sci. USA 81, 
5662-5666 (1984); Zoller, M. J. & Smith, M. Nucleic Acids Research 10, 6487-6500 (1982); Wang, A. et al., Science 
224, 1431-1433; Dalbadie-McFarland, G. etal., Proc. Natl. Acad. Sci. USA 79, 6409-6413 (1982)). In addition, the amino 

25 acid sequences of the antibody constant regions and such are well known to those skilled in the art. 

[0029] In the present invention, the HLA-recognizing minibodies specifically bind to HLA. They are not particularly 
limited, so long as they have a biological action. The minibodies of this invention can be prepared by methods well known 
to those skilled in the art. For example, as described in the Examples, the antibodies can be prepared based on the 
sequence of an HLA-recognizing antibody (particularly sequences of the variable regions and sequences of CDRs), 

30 using genetic engineering techniques known to those skilled in the art. 

[0030] For the sequence of the HLA-recognizing antibody, particularly of the framework region (FR), a well-known 
antibody sequence can be used, or an anti-HLA antibody can be prepared by a method well known to those skilled in 
the art using HLA as the antigen, and then the sequence of this antibody can be obtained and used. Specifically, for 
example, this can be performed as follows: HLA protein, or a fragment thereof, is used as a sensitizing antigen to perform 

35 immunizations according to conventional immunization methods, the obtained immunocytes are fused with well-known 
parent cells according to conventional cell fusion methods, and monoclonal antibody-producing cells (hybridomas) are 
then screened by ordinary screening methods. Antigens can be prepared by known methods, such as a method using 
baculoviruses (W098/46777 and such). Hybridomas can be prepared, for example, according to the method of Milstein 
et al. (Kohler, G. and Milstein, O, Methods Enzymol. (1981) 73:3-46). When the antigen has low immunogenicity, 

40 immunization can be performed using the antigen bound to immunogenic macromolecules, such as albumin. Thereafter, 
cDNAs of the variable region (V region) of the antibody are synthesized from the mRNAs of the hybridomas using reverse 
transcriptase, and the sequences of the obtained cDNAs can be determined by known methods. 
[0031] Antibodies that recognize HLA are not particularly limited, so long as they bind to HLA; mouse antibodies, rat 
antibodies, rabbit antibodies, sheep antibodies, human antibodies, and such may be used as necessary. Alternatively, 

45 artificially modified, genetically recombinant antibodies, such as chimeric and humanized antibodies, may be used to 
reduce heterologous antigenicity against humans. These modified antibodies can be produced using known methods. 
A chimeric antibody is an antibody comprising the variable regions of the heavy and light chains of an antibody from a 
non-human mammal such as a mouse, and the constant regions of the heavy and light chains of a human antibody. 
The chimeric antibody can be produced by linking a DNA encoding the variable regions of the mouse antibody with a 

so DNA encoding the constant regions of the human antibody, incorporating this into an expression vector, and then 
introducing the vector to a host. 

[0032] Humanized antibodies are also referred to as "reshaped human antibodies". Such humanized antibodies are 
obtained by transferring the CDR of an antibody derived from a non-human mammal, for example a mouse, to the CDR 
of a human antibody, and general gene recombination procedures for this are also known. Specifically, a DNA sequence 
55 designed to link a murine antibody CDR to the framework region (FR) of a human antibody can be synthesized by PCR, 
using primers prepared from several oligonucleotides containing overlapping portions of terminal regions. The obtained 
DNA is linked to a DNA encoding human antibody constant regions, and this is then integrated into an expression vector, 
and the antibody is produced by introducing this vector into host cells (see European Patent Application EP 239400, 



6 



EP 1 757 686 A1 



and International Patent Application WO 96/02576). The human antibody FR to be linked via the CDR is selected so 
the CDR forms a favorable antigen-binding site. To form a suitable antigen-binding site in the CDR of the reshaped 
human antibody, amino acids in the framework region of the antibody variable region may be substituted as necessary 
( Sato, K. et al., 1993, Cancer Res. 53, 851-856). 
5 [0033] These chimeric antibodies and humanized antibodies can be chimerized, humanized, and such after their 
molecular weight is reduced, or their molecular weight can be reduced after they have been chimerized, humanized, or 
such. 

[0034] Methods for obtaining human antibodies are also known. For example, human lymphocytes can be sensitized 
in w'frawith a desired antigen, or with cells expressing the desired antigen, and the sensitized lymphocytes can be fused 

10 with human myeloma cells, such as U266, to obtain the desired human antibody with antigen-binding activity (Examined 
Published Japanese Patent Application No. (JP-B) Hei 1-59878). Further, a desired human antibody can be obtained 
by using a desired antigen to immunize transgenic animals that have a full repertoire of human antibody genes (see 
International Patent Application WO 93/12227, WO 92/03918, WO 94/02602, WO 94/25585, WO 96/34096, and WO 
96/33735). Furthermore, techniques for obtaining human antibodies by panning using a human antibody library are also 

15 known. For example, variable regions of human antibodies can be expressed as single chain antibodies (scFvs) on the 
surface of phages using phage display methods, and phages that bind to antigens can be selected. The DNA sequences 
that encode the variable regions of the human antibodies binding the antigens can be determined by analyzing the genes 
of the selected phages. By determining the DNA sequences of the scFvs that bind to the antigens, appropriate expression 
vectors carrying relevant sequences can be produced to yield human antibodies. These methods are already known, 

20 and are detailed in the following publications: WO 92/01 047, WO 92/20791 , WO 93/0621 3, WO 93/1 1 236, WO 93/1 91 72, 
WO 95/01438, and WO 95/15388. 

[0035] Therefore, the minibodies of the present invention can be chimerized, humanized, or such by methods well 
known to those skilled in the art. Such chimeric and humanized antibodies are also included in the minibodies of this 
invention. 

25 [0036] The antibodies of this invention may be conjugate antibodies that are bonded to various molecules, such as 
polyethylene glycol (PEG), radioactive substances, and toxins. Such conjugate antibodies can be obtained by performing 
chemical modifications on the obtained antibodies. Methods for antibody modification are established in this field. The 
term "antibody" in this invention includes such conjugate antibodies. 

[0037] The present invention includes DNAs that encode the antibodies of this invention. This invention also includes 

so DNAs encoding antibodies that hybridize under stringent conditions to the aforementioned DNAs, and have antigen- 
binding capacity and activity. Hybridization techniques (Sambrook, J. et al., Molecular Cloning 2nd ed., 9.47-9.58, Cold 
Spring Harbor Lab. press, 1 989) are well known to those skilled in the art, and hybridization conditions can be selected 
appropriately by those skilled in the art. Such hybridization conditions include, for example, conditions of low stringency. 
Examples of conditions of low stringency include post-hybridization washing in 0.1x SSC and 0.1% SDS at 42°C, and 

35 preferably in O.lxSSC and0.1%SDSat50°C. More preferable hybridization conditions include highly stringent conditions. 
Highly stringent conditions include, for example, washing in 5x SSC and 0.1% SDS at 65°C. In these conditions, the 
higher the temperature, the higher the expectation of efficiently obtaining DNAs with a high homology. However, several 
factors, such as temperature and salt concentration, can influence hybridization stringency, and those skilled in the art 
can suitably select these factors to achieve similar stringencies. 

40 [0038] The DNAs of this invention are used for in vivo and in vitro production of the antibodies of this invention, and 
for other applications, such as gene therapy. The DNAs of this invention may be in any form, so long as they encode 
the antibodies of this invention. More specifically, they may be cDNAs synthesized from mRNAs, genomic DNAs, chem- 
ically synthesized DNAs, or such. Furthermore, the DNAs of this invention include any nucleotide sequence based on 
the degeneracy of the genetic code, so long as they encode the antibodies of this invention. 

45 [0039] The antibodies of this invention can be produced by methods well known to those skilled in the art. More 
specifically, a DNA of an antibody of interest is incorporated into an expression vector. In so doing, the DNA is incorporated 
into the expression vector and expressed under the control of an expression regulatory region such as an enhancer or 
promoter. Next, antibodies can be expressed by transforming host cells with this expression vector. In this regard, 
appropriate combinations of hosts and expression vectors can be used. 

so [0040] The vectors include, for example, M13 vectors, pUC vectors, pBR322, pBluescript, and pCR-Script. In addition 
to the above vectors, for example, pGEM-T, pDIRECT, andpT7can also be used for the subcloning and excision of cDN As. 
[0041] When using vectors to produce the antibodies of this invention, expression vectors are particularly useful. When 
an expression vector is expressed in E. coli, for example, it should have the above characteristics in order to be amplified 
in E. coli. Additionally, when E. coli such as JM109, DH5oc, HB101, or XL1-Blue are used as the host cell, the vector 

55 preferably has a promoter, for example, a lacZ promoter (Ward et al. (1989) Nature 341 :544-546; (1992) FASEB J. 6: 
2422-2427), araB promoter (Better et al. (1988) Science 240:1041-1043), or T7 promoter, to allow efficient expression 
of the desired gene in E. coli. Other examples of the vectors include pGEX-SX-1 (Pharmacia), "QIAexpress system" 
(QIAGEN), pEGFP, and pET (where BL21, a strain expressing T7 RNA polymerase, is preferably used as the host). 



7 



EP 1 757 686 A1 



[0042] Furthermore, the vector may comprise a signal sequence for polypeptide secretion. When producing proteins 
into the periplasm of E. coli, the pelB signal sequence (Lei, S. P. et al. J. Bacteriol. 169:4379 (1987)) may be used as 
a signal sequence for protein secretion. For example, calcium chloride methods or electroporation methods may be 
used to introduce the vector into a host cell. 

5 [0043] In addition to E. coli, expression vectors derived from mammals (e.g., pCDNA3 (Invitrogen), pEGF-BOS (Nucleic 
Acids Res. (1990) 1 8(1 7):5322), pEF, pCDM8), insect cells (e.g., "Bac-to-BAC baculovirus expression system" (GIBCO- 
BRL), pBacPAK8), plants (e.g., pMH1 , pMH2), animal viruses (e.g., pHSV, pMV, pAdexLcw), retroviruses (e.g., pZIPneo), 
yeasts (e.g., "Pichia Expression Kit" (Invitrogen), pNV11, SP-Q01), and Bacillus subtilis (e.g., pPL608, pKTH50) may 
also be used as a vector for producing a polypeptide of the present invention. 

w [0044] In order to express proteins in animal cells such as CHO, COS, and NIH3T3 cells, the vector preferably has a 
promoter necessary for expression in such cells, for example, an SV40 promoter (Mulligan et al. (1979) Nature 277: 
1 08), MMLV-LTR promoter, EFIocpromoter (Mizushima et al. (1 990) Nucleic Acids Res. 1 8:5322), CMV promoter, etc.). 
It is even more preferable that the vector also carry a marker gene for selecting transformants (for example, a drug- 
resistance gene enabling selection by a drug, such as neomycin and G41 8). Examples of vectors with such characteristics 

15 include pMAM, pDR2, pBK-RSV, pBK-CMV, pOPRSV, pOP 13, and such. 

[0045] In addition, to stably express a gene and amplify the gene copy number in cells, CHO cells having a defective 
nucleic acid synthesis pathway can be introduced with a vector containing a DHFR gene (for example, pCHOI) to 
compensate for the defect, and the copy number may be amplified using methotrexate (MTX). Alternatively, a COS cell, 
which carries an SV40 T antigen-expressing gene on its chromosome, can be transformed with a vector containing the 

20 SV40 replication origin (for example, pcD) for transient gene expression. The replication origin may be derived from 
polyoma viruses, adenoviruses, bovine papilloma viruses (BPV), and such. Furthermore, to increase the gene copy 
number in host cells, the expression vector may contain, as a selection marker, an aminoglycoside transferase (APH) 
gene, thymidine kinase (TK) gene, E. coli xanthine guanine phosphoribosyl transferase (Ecogpt) gene, dihydrofolate 
reductase (dhfr) gene, and such. 

25 [0046] Methods for expressing the DNAs of this invention in the bodies of animals include methods of incorporating 
the DNAs of this invention into appropriate vectors and introducing them into living bodies by, for example, a retrovirus 
method, liposome method, cationic liposome method, or adenovirus method. The vectors that are used include adenovirus 
vectors (for example, pAdexIcw), and retrovirus vectors (for example, pZIPneo), but are not limited thereto. General 
genetic manipulations such as inserting the DNAs of this invention into vectors can be performed according to conventional 

30 methods (Molecular Cloning, 5.61 -5.63). Administration to living bodies can be carried out by ex vivo method or in vivo 
methods. 

[0047] Furthermore, the present invention provides host cells into which a vector of this invention is introduced. The 
host cells into which a vector of this invention is introduced are not particularly limited; for example, E. coli and various 
animal cells are available for this purpose. The host cells of this invention may be used, for example, as production 
35 systems to produce and express the antibodies of the present invention, in vitro and in vivo production systems are 
available for polypeptide production systems. Production systems that use eukaryotic cells or prokaryotic cells are 
examples of in vitro production systems. 

[0048] Eukaryotic cells that can be used include, for example, animal cells, plant cells, and fungal cells. Known animal 
cells include: mammalian cells, for example, CHO (J. Exp. Med. (1995)108, 945), COS, 3T3, myeloma, BHK (baby 

40 hamster kidney), HeLa, Vero, amphibian cells such as Xenopus laevis oocytes (Valle, et al. (1981 ) Nature 291 , 358-340), 
or insect cells (e.g., Sf9, Sf21 , and Tn5). CHO cells in which the DHFR gene has been deleted, such as dhfr-CHO (Proc. 
Natl. Acad. Sci. USA (1980) 77, 4216-4220) and CHO K-1 (Proc. Natl. Acad. Sci. USA (1968) 60, 1275), are particularly 
preferable for use as CHO cells. Of the animal cells, CHO cells are particularly favorable for large-scale expression. 
Vectors can be introduced into a host cell by, for example, calcium phosphate methods, DEAE-dextran methods, methods 

45 using cationic liposome DOTAP (Boehringer-Mannheim), electroporation methods, lipofection methods, etc. 

[0049] Plant cells include, for example, Nicotiana tabacum-derived cells known as polypeptide production systems. 
Calluses may be cultured from these cells. Known fungal cells include yeast cells, for example, the genus Saccharomyces, 
such as Saccharomyces cerevisiae; and filamentous fungi, for example, the genus Aspergillus such as Aspergillus niger. 
[0050] Bacterial cells can be used in prokaryotic production systems. Examples of bacterial cells include E. coli (for 

so example, JM109, DH5oc, HB101 and such); and Bacillus subtilis. 

[0051] Antibodies can be obtained by transforming the cells with a polynucleotide of interest, then culturing these 
transformants in vitro. Transformants can be cultured using known methods. For example, DMEM, MEM, RPMI 1640, 
or IMDM may be used as the culture medium for animal cells, and may be used with or without serum supplements such 
as fetal calf serum (FCS). Serum-free cultures are also acceptable. The preferred pH is about 6 to 8 over the course of 

55 culturing. Incubation is typically carried out at a temperature of about 30 to 40°C for about 15 to 200 hours. Medium is 
exchanged, aerated, or stirred, as necessary. 

[0052] On the other hand, production systems using animal or plant hosts may be used as systems for producing 
polypeptides in vivo. For example, a DNA of interest may be introduced into an animal or plant, and the polypeptide 



8 



EP 1 757 686 A1 



produced in the body of the animal or plant is then recovered. The "hosts" of the present invention include such animals 
and plants. 

[0053] When using animals, there are production systems using mammals or insects. Mammals such as goats, pigs, 
sheep, mice, and cattle may be used (Vicki Glaser SPECTRUM Biotechnology Applications (1993)). Alternatively, the 

5 mammals may be transgenic animals. 

[0054] For example, a DNAof interest may be prepared as a fusion gene with agene encoding a polypeptide specifically 
produced in milk, such as the goat p-casein gene. DNA fragments containing the fusion gene are injected into goat 
embryos, which are then introduced back to female goats. The desired antibody can then be obtained from milk produced 
by the transgenic goats born from the goats that received the embryos, or from their offspring. Appropriate hormones 

10 may be administered to increase the volume of milk containing the polypeptide produced by the transgenic goats (Ebert, 
K.M. etal., Bio/Technology 12, 699-702 (1994)). 

[0055] Insects, such as silkworms, may also be used. Baculoviruses carrying a DNA of interest can be used to infect 
silkworms, and the antibody of interest can be obtained from their body fluids (Susumu, M. et al., Nature 315, 592-594 
(1985)). 

15 [0056] When using plants, tobacco can be used, for example. When tobacco is used, a DNA of interest may be inserted 
into a plant expression vector, for example, pMON 530, and then the vector may be introduced into a bacterium, such 
as Agrobacterium tumefaciens. The bacteria are then used to infect tobacco such as Nicotiana tabacum, and the desired 
polypeptides are recovered from the leaves (Julian K.-C. Ma et al., Eur. J. Immunol. 24,131-138(1994)). 
[0057] The resulting antibodies of this invention may be isolated from the inside or outside (such as the medium) of 

20 host cells, and purified as substantially pure and homogenous antibodies. Any standard method for isolating and purifying 
antibodies may be used, and methods are not limited to any specific method. Antibodies may be isolated and purified 
by selecting an appropriate combination of, for example, chromatographic columns, filtration, ultrafiltration, salting out, 
solvent precipitation, solvent extraction, distillation, immunoprecipitation, SDS-polyacrylamide gel electrophoresis, iso- 
electric focusing, dialysis, recrystallization, and others. 

25 [0058] Chromatography includes, for example, affinity chromatography, ion exchange chromatography, hydrophobic 
chromatography, gel filtration, reverse-phase chromatography, and adsorption chromatography (Strategies for Protein 
Purification and Characterization: A Laboratory Course Manual. Ed Daniel R. Marshak et al., Cold Spring Harbor Lab- 
oratory Press, 1996). These chromatographies can be carried out using liquid phase chromatographies such as HPLC 
and FPLC. The present invention also includes antibodies that are highly purified using these purification methods. 

30 [0059] In the present invention, the antigen-binding activity of antibodies (Antibodies A Laboratory Manual. Ed Harlow, 
David Lane, Cold Spring Harbor Laboratory, 1 988) can be measured using well known techniques. For example, ELISA 
(enzyme linked immunosorbent assay), EIA (enzyme immunoassay), RIA (radioimmunoassay), or fluoroimmunoassay 
may be used. 

[0060] In the present invention, whether or not the antibodies of this invention induce cell death in non-adherent cells 
35 can be determined from whether cell death is induced in Jurkat cells or ARH77 cells, as in the Examples. Whether or 
not the antibodies induce cell death in adhesion cells can be determined from whether cell death is induced in HeLa 
cells, as in the Examples. 

[0061 ] Furthermore, the present invention provides cell death-inducing agents or cell growth-suppressing agents which 
comprise minibodies or 2D7 antibodies of this invention as active ingredients. The cell death-inducing activity of the 

40 minibodies or 2D7 antibodies in this invention is considered to have a particularly large effect on activated T cells or B 
cells; therefore, it is considered to be particularly effective for treatment and prevention of tumors such as cancer 
(particularly blood tumors), and autoimmune diseases. Accordingly, the present invention provides methods of treatment 
and prevention of tumors such as cancer (particularly blood tumors), and autoimmune diseases that use the minibodies 
or 2D7 antibodies of this invention. When using 2D7 antibodies whose molecular weight has not been reduced as active 

45 ingredients, they are preferably cross-linked with an anti-IgG antibody and such. 

[0062] The above-mentioned antibodies can also be used as conjugate antibodies, after linking to various reagents. 
Examples of such reagents include chemotherapy reagents, radioactive substances, and toxins. Such conjugate anti- 
bodies can be produced by known methods (US5057313, and US51 56840). 

[0063] The above-mentioned pharmaceutical agents can be directly administered to patients, or administered as 
so pharmaceutical compositions formulated by known pharmaceutical methods. For example, they may be administered 
orally, as tablets, capsules, elixirs, or microcapsules, sugar-coated as necessary; or parenteral ly, in the form of injections 
of sterile solution or suspensions prepared with water or other pharmaceutical^ acceptable liquids. For example, they 
may be formu lated by appropriately combin ing them with pharmaceutical^ acceptable carriers or media, more specifically, 
sterilized water or physiological saline solutions, vegetable oils, emulsifiers, suspending agents, surfactants, stabilizers, 
55 flavoring agents, excipients, vehicles, preservatives, binding agents, and such, and mixing them at a unit dosage form 
required for generally accepted pharmaceutical practice. The amount of active ingredient in the formulation is such that 
appropriate doses within indicated ranges are achieved. 

[0064] Additives that can be mixed into tablets and capsules include, for example, binding agents such as gelatin, 



9 



EP 1 757 686 A1 



cornstarch, tragacanth gum, and gum arabic; excipientssuch as crystalline cellulose; swelling agents such as cornstarch, 
gelatin, alginic acid; lubricants such as magnesium stearate; sweeteners such as sucrose, lactose, or saccharine; and 
flavoring agents such as peppermint and Gaultheria adenothrix oils, or cherry. When the unit dosage form is a capsule, 
liquid carriers, such as oils and fats, can be further included in the above-indicated materials. Sterile compositions to be 

5 injected can be formulated using a vehicle such as distilled water used for injection, according to standard protocols. 
[0065] Aqueous solutions used for injections include, for example, physiological saline and isotonic solutions com- 
prising glucose or other adjunctive agents such as D-sorbitol, D-mannose, D-mannitol, and sodium chloride. They may 
also be combined with appropriate solubilizing agents, such as alcohol, and specifically, ethanol, polyalcohol such as 
propylene glycol or polyethylene glycol, or non-ionic detergent such as polysorbate 80™ or HCO-50, as necessary. 

w [0066] Oil solutions include sesame oils and soybean oils, and can be combined with solubilizing agents such as 
benzyl benzoate or benzyl alcohol. Injection solutions may also be formulated with buffers, for example, phosphate 
buffers or sodium acetate buffers; analgesics, for example, procaine hydrochloride; stabilizers, for example, benzyl 
alcohol or phenol; or anti-oxidants. The prepared injections are typically aliquoted into appropriate ampules. 
[0067] Administration to patients may be performed, for example by intra-arterial injection, intravenous injection, or 

15 subcutaneous injection, alternatively by intranasal, transbronchial, intramuscular, transdermal, or oral administration 
using methods well known to those skilled in the art. Doses vary depending on the body weight and age of the patient, 
method of administration and such; nevertheless, those skilled in the art can appropriately select suitable doses. Fur- 
thermore, if a compound can be encoded by a DNA, the DNA may be incorporated into a gene therapy vector to carry 
out gene therapy. Doses and administration methods vary depending on the body weight, age, and symptoms of patients, 

20 but, again, they can be appropriately selected by those skilled in the art. 

[0068] A single dose of a pharmaceutical agent of this invention varies depending on the target of administration, the 
target organ, symptoms, and administration method. However, the usual dose for an adult (presuming a body weight of 
60 kg) in the form of an injection is approximately 0.1 to 1000 mg, preferably approximately 1.0 to 50 mg, and more 
preferably approximately 1 .0 to 20 mg per day, for example. 

25 [0069] When administered parenterally, a single dose varies depending on the target of administration, the target 
organ, symptoms, and administration method. However, when in the form of an injection to an adult (presuming a body 
weight of 60 kg), usually it is considered advantageous to intravenously administer, for example, a single dose of about 
0.01 to 30 mg, preferably about 0.1 to 20 mg, and more preferably about 0.1 to 10 mg per day. For other animals, a 
converted amount based on the amount for a body weight of 60 kg, or a converted amount based on the amount for a 

so body surface area can be administered. 

Brief Description of the Drawings 
[0070] 

35 

Fig. 1 shows the adaptors used to produce the pMX2 vector. The bold letters indicate BstXI recognition sequences. 
Fig. 2A and Fig. 2B show 2D7 antigen expression in cell lines. Each cell type was stained with 2D7 antibody and 
the expression examined. (Solid line: no primary antibody; dotted line: 2D7 antibody) 

Fig. 3 is a set of photographs showing the results of immunoprecipitation using the 2D7 antibody. NIH3T3, RPMI 
40 8226, and U266 cells were solubilized, immunoprecipitation was performed with the 2D7 antibody, anti-BST-1 

antibody (control), or protein G itself, and the proteins were detected by silver staining. In RPMI 8226 and U266, a 
molecule of approximately 12 KD (arrow), which is specifically precipitated by the 2D7 antibody, was detected. This 
band was cut out and peptide sequenced, and thus found to be J32-microglobulin. 

Fig. 4 shows flow diagrams for screening. Separation into pools, preparation of DNA, packaging into virus, infection 
45 of 3T3 cells, and screening using FACS were performed in one span (Fig. 4A). By the end of the fourth screening, 

the library was narrowed down to approximately 20 clones. In the fifth screening, 64 colonies were individually 
inoculated into a 96-well plate, pools were formed using the vertical and horizontal rows, and then screened. As a 
result, the library was narrowed down to twelve candidate clones (Fig. 4B). 

Fig. 5 shows the results of screening using FACS. Fig. 5A shows the results of the second screening, Fig. 5B shows 
so the results of the third screening, and Fig. 5C shows the results of the fourth screening. NIH3T3 cells were infected 

with retroviruses prepared from each pool, and three days later the cells were stained with the 2D7 antibody. The 
clones were narrowed down by gradually reducing the pool size of each screening. 

Fig. 6 shows the results of screening using FACS. Fig. 6A shows the results of the fifth screening, and Fig. 6B shows 
the result of the final screening. As a result of the fifth screening, positive clones were found in rows 3, 4, 6, and 8, 
55 and in rows E, F, and G. As a result of screening the twelve candidate clones, positive clones were found in row E 

at 6E . When the nucleotide sequence of this 6E was analyzed, it was found to encode HLA classl A*6802. 
Fig. 7 is a graph and a set of photographs showing the influence on cells of the addition of 2D7 antibody. 2D7 
antibody (1 0 |j.g/ml) was added, and the number of viable cells was determined 48 hours later. Hardly any change 



10 



EP 1 757 686 A1 



in cell growth was observed, even after 2D7 antibody was added (Fig. 7A). K562 cells (Fig. 7B), Jurkat cells (Fig. 
7C), and RPMI8226 cells (Fig. 7D) were each observed 24 hours after 2D7 antibody addition. The 2D7 antibody 
induced aggregation of Jurkat cells. 

Fig. 8 is a set of photographs showing cell death induction due to cross-linking of the 2D7 antibody. Each combination 
5 of the 2D7 antibody with anti-mouse IgG was made to act on Jurkat cells, and the cell nuclei were stained 48 hours 

later. Nuclear fragmentation due to cell death was observed when the 2D7 antibody and anti-mouse IgG acted on 
cells simultaneously. 

Fig. 9 shows a 2D7 diabody (2D7DB) sequence. The nucleotide sequences in the figure is shown in SEQ ID NO: 
3, and the amino acid sequence is shown in SEQ ID NO: 4. 
10 Fig. 10A and Fig. 10B show a 2D7 diabody structure. Fig. 10C is a photograph showing 2D7 diabody transient 

expression in COS7 cells. 

Fig. 1 1 A and Fig. 1 1 B show the cytotoxic activity of 2D7DB transiently expressed in COS7. 

Fig. 12 shows the cytotoxic activity of 2D7DB transiently expressed in COS7. K562 cells (Fig. 1 2A) and Jurkat cells 
(Fig. 12B) were used. 

15 Fig. 13 shows the cytotoxic activity of 2D7DB transiently expressed in COS7. RPMI 8226 cells (Fig. 13A), IL-KM3 

cells (Fig. 13B), U266 cells (Fig. 13C), and ARH77 cells (Fig. 13D) were used. 
Fig. 14 is a graph showing the growth-suppressing effect of purified 2D7DB. 

Fig. 15 shows cell death induction by purified 2D7DB, 48 hours after induction. ARH77 cells (Fig. 15A), Jurkat cells 
(Fig. 15B), K562 cells (Fig. 15C), and HeLa cells (Fig. 15D) were used. 
20 Fig. 16 shows cell death induction by purified 2D7DB, 48 hours after induction. U266 cells (Fig. 16A), and IL-KM3 

cells (Fig. 1 6B) were used for the study. 

Fig. 17 shows a time course of cell death induction by 2D7DB (2 |j,g/ml). Cell death induction was investigated at 
12 through to 38 hours. ARH77 cells (Fig. 17A) and Jurkat cells (Fig. 17B) were used. 

Fig. 18 shows a time course of cell death induction by 2D7DB (2 |j,g/ml). Cell death induction was investigated at 
25 three through to six hours. ARH77 cells (Fig. 1 8A) and Jurkat cells (Fig. 1 8B) were used. 

Fig. 19 shows the effect of Z-VAD-FMK on cell death due to 2D7DB. The study was performed using ARH77 cells 
16 hours after induction. 

Fig. 20 shows the effect of Z-VAD-FMK on cell death due to 2D7DB. The study was performed using Jurkat cells 
16 hours after induction. 

30 Fig. 21 is a set of photographs showing that cell death due to 2D7DB is not accompanied by DNA fragmentation. 

The study was performed 24 hours after cell death induction. 

Fig. 22 shows the results of investigating the effect of cytochalasin D on the cell death-inducing activity of 2D7DB. 
By pre-treating ARH77 cells with cytochalasin D, which is an actin-polymerization inhibitor, the cells showed resist- 
ance to 2D7DB-induced cell death. 
35 Fig. 23 is a set of photographs showing the results of immunostaining to investigate the state of the intracellular 

actin and nuclei. After reacting ARH77 cells under the conditions described in the figure, actin was detected using 
anti-actin antibody (red), and cell nuclei were detected using Hoechst 33258 (blue). Actin was absent in cells treated 
with 2D7DB. 

Fig. 24 shows that 2D7DB suppresses an increase in human IgG (hlgG) concentration in serum in a mouse model 
40 of human myeloma. The data shows the average + SEM. There was a significant difference (*: p<0.05) between 

the vehicle-administered group and the 2D7DB-administered group, according to unpaired t-tests. 
Fig. 25 shows that 2D7DB has a life-prolonging effect in a mouse model of human myeloma. There was a significant 
difference (*: p<0.05) between the vehicle-administered group and the 2D7DB-administered group, according to 
generalized Wilcoxon tests. 

45 Fig. 26 shows analyses of the action of 2D7DB on PBMC. PHA-M (Fig. 26A), ConA (Fig. 26B), and SAC (Fig. 26C) 

were used as mitogens. Fig. 26D shows the results in the absence of a mitogen, and Fig. 26E shows the results of 
a positive control (ARH77). The results shown are, from the top, those of no 2D7DB addition, three-hour addition, 
and 24-hour addition. 

so Best Mode for Carrying out the Invention 

[0071] Herein below, the present invention is specifically described using Examples; however, it should not to be 
construed as being limited thereto. 

55 [1] Cell lines 

[0072] Human myeloma cell lines (RPMI8226, K562, and ARH77), human T-cell leukemia cell line (Jurkat), FDC-P1 , 
HCI-16, and 2D7 hybridoma cell line (from University of Tokushima) were cultured in RPMI1640 medium (GIBCO BRL) 



11 



EP 1 757 686 A1 



supplemented with 1 0% fetal calf serum (FCS). Human myeloma cell lines (IL-KM3 and U266) were individually cultured 
in the same medium supplemented with 2 ng/ml of IL-6 (R&D), and Ba/F3 was cultured in the same medium supplemented 
with 2 ng/ml of IL-3 (R & D). COS7, 293T, HeLa, NIH3T3, and BOSC23 were cultured in DMEM medium (GIBCO BRL) 
supplemented with 10% FCS, and CHO was cultured in oc-MEM medium (GIBCO BRL) supplemented with 5% FCS or 
5 10% FCS. 

[2] Production of pMX2 vectors 

[0073] The GFP gene region of the retrovirus vector, pMX-GFP, which packages the GFP gene into the virus particle, 
10 was cut out and removed using EcoRI-Sall. The adaptor, which comprised a BstXI site in its sequence (Fig. 1 ) (and was 
synthesized with anABI DNA synthesizer, then annealed in vitro before use), was inserted into this region, forming pMX2. 

[3] Production of cDNA libraries 

15 [0074] Total RNA was purified from RPMI8226 cells by standard methods using TRIzol (GIBCO BRL). Furthermore, 
the mRNAs were purified from 200 |j,g of this total RNA, using &\x, MACS mRNA Isolation kit (Miltenyi Biotec) according 
to the manufacturer's instructions. The cDNAs were synthesized using random hexamers (Superscript Choice System 
for cDNA Synthesis; Invitrogen) with 3.6 |j.g of mRNA as template, and then a BstXI adaptor (Invitrogen) was linked to 
both ends. This cDNA was inserted into the pMX2 vector cleaved with BstXI, and was introduced into ELECTRO MAX 

20 DH10B (GIBCO BRL) by electroporation (2.5 KV, 200 O, 25 |xF). After adding 1 ml of SOC, the vectors were then 
incubated at 37°C for one hour, 1 ml of 40% glycerol/LB+Amp was added. A portion of the culture was used to check 
the titer and the remainder was stored at -80°C. The obtained library was plated at 200|j,l/well (7% DMSO/LB+Amp) 
into two 96-well plates, so that each well contained 1000 clones. These were cultured overnight at 37°C. Four wells 
(4000 clones) from this plate were combined and placed into an ampicillin-containing LB medium (4 ml). This was defined 

25 as one pool, the rest of the wells were treated similarly. Ultimately, 24 pools were prepared from a single plate. After 
incubating each pool overnight at 37° C, DNAs were prepared (QIAGEN) and used for transfection into packaging cells. 
The plates used for inoculation were stored at -80 °C until use in secondary screening. 

[4] Purification of antibodies 

30 

[0075] 0.5 ml of ascites, sent from University of Tokushima, was adsorbed to a Protein A Hi Trap Affinity column 
(Amersham Pharmacia). The IgG fraction was then eluted using 0.1 M sodium citrate, pH3.0, and the 2D7 antibody was 
collected. This was concentrated using Centricon (YM-1 0; Millipore), and the buffer was exchanged to PBS to ultimately 
yield a total of 5.34 mg of antibody. This was separated into aliquots and stored at -20°C (concentration: 0.89|j,g/|jiL). 

35 

[5] FACS 

[0076] Adherent cells were detached using 1 mM EDTA/PBS, and non-adherent cells were collected by centrifugation, 
then suspended in FACS buffer (2.5% FCS, 0.02% NaN 3 /PBS). These cells were left to stand on ice for one hour in a 
40 buffer (5% FCS/PBS) containing 2D7 antibody (final concentration 10 |j.g/ml). These were then washed with FACS 
buffer, reacted in a solution of FITC-anti-mouse IgG (Immunotech) (1:150, 50 jjlL FACS buffer) on ice for 30 minutes, 
washed twice with FACS buffer, and then analyzed using EPICS ELITE (COULTER). 

[6] Retrovirus infection 

45 

(i) Retrovirus packaging 

[0077] The day before transfection, 2ml of BOSC23 cells, which are retrovirus-packaging cells, were plated onto a 6- 
well plate at 6 x 1 0 5 cells/well. Transfection was carried out by the following procedure: 1 |j.g of the plasmid DNA derived 
so from each pool was mixed with 3 |j.L of FuGENE 6 Transfection Reagent (Roche), left to stand at room temperature for 
20 minutes, and then added to the BOSC23 cell culture medium plated the day before. Cells were then cultured at 37 °C 
for 48 hours, and the culture medium was collected. Dead cells were removed by centrifugation at 3000 rpm for five 
minutes, and the culture solution was then used as the virus solution. 

55 (ii) Virus infection 

[0078] NIH3T3 cells plated onto 6-well plates (1 x 1 0 5 cells/2ml/well) the day before virus infection. Then 1 ml of virus 
solution supplemented with 1 0 |j,g/ml of polybrene (hexadimethrine bromide; Sigma) was added to the wells and NIH3T3 



12 



EP 1 757 686 A1 



cells were cultured for 24 hours. 1 .5 ml of fresh medium was then added, the cells were cultured for another 48 hours, 
and gene expression was then analyzed using FACS. 

[7] Immunoprecipitation 

5 

[0079] Cells were lysed in a lysis buffer (0.5% Nonidet P-40, 1 0 mM Tris, pH 7.6, 150 mM NaCI, 5 mM EDTA, 1 mM 
phenylmethylsulfonyl fluoride, 5 |j.g/ml aprotinin), and the resulting solution was centrifuged to remove the insoluble 
proteins and obtain a cell lysate. 1 |j,g of 2D7 antibody was added, and incubated at 4°C for four hours. Magnetic protein 
G (BioMag) was then added, and this was incubated for another one hour. Subsequently, the immunoconjugate was 

10 washed three times with a lysis buffer, and then subjected to SDS-PAGE. This gel was silver stained (Daiichi Pure 
Chemicals) according to the attached instructions. For peptide sequencing, the gel on which SDS-PAGE was performed 
was transferred to ProBlott (Applied Biosystems), and this was stained for one minute with Coomassie blue staining 
solution (0.1% coomassie blue R-250 in 40% MetOH/1% acetic acid). After washing several times with 50% MetOH, 
the band of interest was cut out, washed five times with 1 ml of DDW, dried in vacuo, and then subjected to peptide 

15 sequencing. 

[8] Cell growth assay using the 2D7 antibody 

[0080] Each type of cell was plated into a 96-well plate at 1 x 10 s cells/ml in the presence or absence of PMA (50 
20 ng/ml; GIBCO BRL) and PHA(10 (xl/ml; GIBCO BRL). This was cultured for 48 hours after subsequent addition (10 
|j,g/ml) or no addition of the 2D7 antibody,. After culturing, morphological changes in the cells were observed under a 
microscope. The relative viable cell count was determined by adding WST-8 (viable cell count reagent SF; Nacalai 
Tesque), culturing at 37 °C for two hours, and measuring OD 450 . 

25 [9] Induction of cell death by cross-linking 

[0081] Jurkat cells were plated on a 24-well plate at 8 x 1 0 5 celis/well, and 1 0 |j,g/ml of anti-mouse IgG (Fc) antibody 
(Cappel) was further added in the presence (5 |j.g/ml) or absence of 2D7 antibody. 48 hours later, the cells were collected, 
and after washing with PBS, methanol was added to a concentration of 70%, and this was left to stand at -20° C for 1 5 
30 minutes. After washing the cells several times with FACS buffer, Hoechst 33258 was added at a concentration of 10 
|j,g/ml, and this was incubated at room temperature for 30 minutes. The cells were washed again with FACS Buffer, and 
an aliquot of the cells was then placed on a slide glass to observe the state of the nuclei under a fluorescence microscope. 

[1 0] Cloning of the 2D7 variable region 

35 

[0082] Total RNA was purified from 2D7 hybridoma (provided by University of Tokushima) using TRIzol according to 
standard methods. Using 3 |j,g of this RNA as a template, cDNAs were synthesized using a SMART RACE cDNA 
Amplification kit (CLONTECH), according to the attached instructions. Using this cDNA as a template, the variable 
regions of the heavy chain and light chain were amplified by PCR using the following primers: 

40 

Heavy chain: 5'-CAGGGGCCAGTGGATAGACTGATG (SEQ ID NO: 7) 
Light chain: 5'-GCTCACTGGATGGTGGGAAGATG (SEQ ID NO: 8) 

The amplified cDNAs encoding each of variable regions were subcloned into pCR-TOPO vector (Invitrogen), and 
the nucleotide sequences (SEQ ID NOs: 1 and 2) were determined. 

45 

[1 1] Production of 2D7 diabody expression vector 

[0083] Plasmids, to which each of the variable region cDNAs were subcloned, were used as templates, and the variable 
regions of the heavy chain and light chain (VH and VL) were respective amplified using the primers below: 

50 

Heavy chain 

2D7DB-H1 : 5'-CCTGAATTCCACCATGCGATGGAGCTGGATCTTTC (SEQ ID NO: 9) 

2D7DB-H2: 5'-AATTTGGCTACCGCCTCCACCTGAGGAGACTGTGAGAGTGGTGCCCT (SEQ ID NO: 10) 

Light chain 

55 2D7DB-L1 : 5'-TCCTCAGGTGGAGGCGGTAGCCAAATTGTTCTCACCCAGTCGCCAGC (SEQ ID NO: 1 1 ) 



13 



EP 1 757 686 A1 



2D7DB-L2: 

5 ' -ATTGCGGCCGCTTATCACTTATCGTCGTCATCCTTGTAGTCTTTTATCTCCAACTTTG 
s TCCCCGAGCC (SEQ ID NO: 12) 

[0084] Each of the VH and VL cDNAs amplified by these primers were combined into one tube, and further subjected 
to PCR. Using the PCR products as templates, PCR was performed again, this time using 2D7DB-H1 and 2D7DB-L2 
as primers, to synthesize cDNAwith VH and VL linked through a 5-mer linker (SEQ ID NO: 3). This cDNA was digested 
10 with EcoRI-Notl and inserted into the EcoRI-Notl gap of the animal cell expression vector, pCXND3. The nucleotide 
sequence was confirmed, completing the construction of the 2D7 diabody expression vector, pCXND3-2D7DB. 

[12] Transient expression in COS7 cells 

15 [0085] 2 |j,g of pCXND3-2D7DB, or of an empty vector used as a control, was mixed with 6 |mL of transfection reagent 
(LT-1, MIRUS) according to the attached instructions, and this was added to COS7 cells (plated the day before into a 
6-well plate at 1x 10 5 cells/well) whose medium had been exchanged to a serum-free medium (OPTI-MEM, GIBCO 
BRL). Five hours later, 200 |jlL of serum was added, and this was cultured for two to three days. The medium was 
collected, and dead cells were removed by centrifugation. The culture supernatant was then used for an experiment to 

20 detect cytotoxic activity. 

[0086] Expression of 2D7DB in the culture supernatant was confirmed by Western blotting. More specifically, equal 
amounts of 2x SDS-PAGE Sample buffer and culture supernatant were added. In addition, after lysing the cells by adding 
a lysis buffer (0.5% Nonidet P-40, 1 0 mM Tris, pH 7.6, 1 50 mM NaCI, 5 mM EDTA), insolubilized proteins were removed 
by centrifugation to prepare a cell lysate, and an equal amount of 2x SDS-PAGE Sample buffer was added to this. After 

25 performing SDS-PAGE on each sample, the gels were transferred to PVDF membranes, and expression of the 2D7 
single chain was detected using anti-FLAG antibody. 

[13] Establishment of expression cell lines producing 2D7 diabody 

30 [0087] 20 fig of pCXND3-2D7DB, linearized by cleaving with Pvul, was introduced to CHO cells (DXB11 strain) by 
electroporation, as described below. 

[0088] After washing the CHO cells twice with ice-cold PBS, they were suspended in PBS at 1x 10 7 cells/ml. 20 |jig 
of the above-mentioned plasmid was mixed into these cells, and this was electropulsed (1.5 KV, 25 ijiFD). The cells 
were diluted into appropriate fractions, plated on to a 10 cm dish, and cultured in the presence of G418 (GIBCO BRL) 
35 at a fmal concentration of 50 |j,g/ml. Approximately 30 clones were selected from the grown colonies, and the diabody 
expression levels in the culture supernatants were investigated by Western blotting. The clone with the highest expression 
level was expanded in a nucleic acid-free MEMoc medium containing 5 nM MTX, and this was stocked as a high-producing 
cell line. 

40 [14] Large-scale purification of 2D7 diabodies 

[0089] A subconfluent 2D7DB-producing CHO cell line in a T-125 flask was detached using Trypsin-EDTA, and then 
this was transferred to a roller bottle (250 ml of MEM a without nucleotide + 5% FCS). Four days later, the culture solution 
was removed, and the cells were washed twice with PBS. The medium was then exchanged to 250 ml of CHO-S-SFMII 

45 medium (GIBCO BRL) to produce a serum-free medium, cells were cultured for three days, and then the cell culture 
supernatant was collected. This was filtered and used for purification after removing the dead cells by centrifugation,. 
[0090] Purification of single chain Fv was performed as follows: First, the collected culture supernatant was applied 
and adsorbed onto an anti-Flag M2 column. After washing with buffer A (50 mM Tris-HCI pH7.4, 150 mM NaCI, 0.01% 
Tween 20), single chain Fv was eluted with buffer B (100 mM Glycine pH3.5, 0.01% Tween 20). The collected sample 

so was immediately neutralized with Tris-HCI pH8.0 so that the final concentration was 25 mM. This was then used for gel 
filtration purification by a Superdex 200HR (26/60) column. The dimer fraction of single chain Fv was collected in PBS 
containing 0.01 % Tween 20. A portion of the collected sample was subjected to SDS electrophoresis and silver staining 
to confirm that the protein of interest has been purified, and then this was concentrated to produce a purified 2D7 diabody 
preparation. 

55 

[15] Cell death induction experiment using 2D7 diabody 

[0091] Various blood cell lines were plated into 24-well plates at 2-5 x 10 5 cells/well. Purified 2D7DB, or the culture 



14 



EP 1 757 686 A1 



supernatant of COS7 transiently expressing 2D7DB, was added and cell death was induced. When the cultu re supernatant 
of COS7 transiently expressing 2D7DB was used, the supernatant was added till its concentration was 50%. The amount 
of medium in each well was 0.8 to 1 ml/well. When stimulating Jurkat cells, Con A (WAKO) was added at the time of 
2D7DB addition to a final concentration of 2 |j,g/ml. 
5 [0092] Adherent cells (HeLa) were plated into a6-well plate at 2x 1 0 5 cells/well, and the cells were attached by culturing 
overnight. Subsequently, purified 2D7DB was added to the culture solution. 

[0093] Several hours to several days after 2D7DB addition, the non-adherent cells were collected as they were, and 
adherent cells were collected after detaching the cells with 1 mM EDTA/PBS. The cells were then washed with ice-cold 
PBS, and labeled with Annexin V, which is an apoptosis marker, and with PI, which is a dead-cell marker, according to 
10 the attached instructions (TACS Annexin V-FITC Apoptosis Detection Kit, TREVIGEN Instructions). The proportion of 
stained cells was then measured using flow cytometry (EPICS ELITE, COULTER). 

[16] Cell death induction by Actinomycin D 

15 [0094] Various blood cell lines were plated into 24-well plates at 2-5 x 10 5 cells/well. To inhibit the initial stage of 
apoptosis, acaspase inhibitor (Z-VAD-FMK, Promega) was added at afinal concentration of 50 |xM, and after incubating 
for 2.5 hours, cell death was induced. For cell death induction by Actinomycin D, Actinomycin D (Sigma) was added at 
1 |j.g/ml (Jurkat) or 5 |j.g/ml (ARH77), and for cell death induction by 2D7DB, 2 |j.g/ml of purified 2D7DB was added. 
Cells were collected 1 6 hours after cell death induction, and stained using Annexin V and PI. 

20 

[1 7] Cell growth assay using 2D7 diabody 

[0095] Each type of cells was plated into a 96-well plate at a cell concentration of 1-2 x 10 4 cells/well. 2D7DB was 
added at an appropriate concentration, and the cell count was determined after three days of culturing. Viable cell count 
25 was determined using WST-8. More specifically, this reagent was added to the cells at 10 |xl/well, and the cells were 
then cultured at 37 °C for 1.5 hours. The relative viable cell count was determined by measuring the OD 450 using a 
spectrophotometer. The growth suppression rate was calculated from (1- (OD 450 of 2D7DB treated cells / OD 450 of 
2D7DB untreated cells)) x 100. 

30 [1 8] Detection of DNA fragmentation 

[0096] ARH77 and Jurkat cells were plated into a 6-well plate so that the cell concentration was 2 x 10 s cells/well, 
and cell death was induced by adding purified 2D7DB at a final concentration of 2 |j.g/ml, or Actinomycin D at a final 
concentration of 1 |j,g/ml (ARH77) or 5 |j,g/ml (Jurkat) to each well. The control was a well to which nothing was added. 
35 After culturing for 24 hours, the cells were collected, washed once with PBS, and then lysed in a lysis buffer (10 mM 
Tris pH7.5, 1 0 mM EDTA, 0.5% Triton X-1 00). This was followed by centrifugation to remove the insoluble proteins, and 
then the material was treated with RNase A and Proteinase K. A portion of this was then subjected to agarose gel 
electrophoresis to detect chromatin DNA fragmentation. 

40 [1 9] Inhibition of cell death induction by cytochalasin D 

[0097] ARH77 cells were plated into a 24-well plate to achieve a cell concentration of 5 x 1 0 5 cells/well, and cytochalasin 
D (Sigma) was added to a final concentration of 20 |j,g/ml. The control was a well to which ethanol alone was added. 
After culturing for one hour, purified 2D7DB was added at various concentrations (0, 200, 500, 1 000 ng/ml), and culturing 
45 was continued for another four hours. Cells were then collected, and the proportion of dead cells was detected by staining 
with PI. 

[20] Immunostaining of 2D7DB-treated cells using anti-actin antibody 

so [0098] 2D7DB was added at a concentration of 1 |j.g/ml to cytochalasin D-treated/-untreated ARH77 cells, and after 
culturing at 37 °C for 15 minutes, the cells were adhered to a slide glass with a Cytospin. After immobilizing the cells by 
immersion in methanol for 15 minutes at -20°C, blocking was performed using a blocking buffer (3% BSA/PBS) at 4°C 
for one hour. This was then reacted with CY3-labeled anti-actin antibody (Sigma) diluted 1 00-fold in 1% BSA/PBS for 
one hour at room temperature, and then the cell nuclei were stained with Hoechst 33258. After washing several times 

55 with PBS, the cells were observed under a confocal laser scanning microscope (Olympus). 



15 



EP 1 757 686 A1 



[Example 1] Expression analysis of 2D7 antigen in each type of cell line 

[0099] To determine the cell line that should become the source to produce a cDNA expression library and the cell 

line that should become the host, 2D7 antigen expression in each type of animal cell was analyzed using FACS (Fig. 
5 2A and Fig. 2B). As a result, among human-derived blood cells, extremely strong expression of the 2D7 antigen was 

observed in lymphocytic tumor cell lines, RPMI8226, U266, and in Jurkat, but expression was found to be weak in K562. 

In Ba/F3, FDC-P1, and HCI-16, which are blood cells derived from mice, expression was very weak, perhaps due to 

differences between species. Of the adherent cells, expression was observed in COS7, 293T, and HeLa. Expression 

was hardly observed in mouse NIH3T3 cells. 
10 [0100] From the expression patterns mentioned above, RPMI8226 cells were judged to be appropriate as a cDNA 

library source to be used for expression cloning, and NIH3T3 cells were determined to be appropriate as host cells to 

be used for screening, to which the expression library is transferred. 

[Example 2] Cloning of 2D7 antigen 

15 

[1] Cloning from a protein 

[0101] Cell lysates were prepared from RPMI8226 cells and U266 cells, which express the 2D7 antigen, and NIH3T3 
cells, which do not express the 2D7 antigen, and immunoprecipitation was performed using the 2D7 antibody. As a 
20 result, a molecule (approximately 12 kD) that precipitates specifically in RPMI8226 and U266 cells was observed (Fig. 
3). This molecule was not detected by Western blotting using the 2D7 antibody, but since it is at least reproducibly 
precipitated by the 2D7 antibody, it was strongly predicted to be the 2D7 antigen itself, or a molecule that co-precipitates 
with the 2D7 antigen. 

[0102] Coomassie staining was performed on this band; it was then cut out and the peptides were sequenced. As a 
25 result, this 12 kD molecule was identified as [32 microglobulin ([}2M). Since [32M is one of the class I MHC protein 
complexes that associate with HLA class I through non-covalent bonds, the 2D7 antibody is considered to have co- 
precipitated it as an HLA complex. HLA class I comprises the oil and oc2 domains required for antigen presentation, and 
the oc3 domain which binds to p2M. Since the 2D7 antibody can co-precipitate the p2M molecule, it is anticipated that 
the 2D7 antibody will recognize the oc1-cc2 domains of HLA class I as an epitope. 

30 

[2] Expression cloning of genes 

[0103] cDNAs were synthesized using random hexamers from mRNAs purified from the 2D7 antigen-expressing cells, 
RPMI8226. These were inserted into a retrovirus vector, pMX2, and a retrovirus expression library was constructed. 
35 The library titer was investigated, and found to include a total of 6 x 1 0 s clones. Furthermore, the average cDNA length 
was found to be approximately 1 .5 kb, arrived at by randomly selecting 24 clones from this library and investigating their 
insert size using colony PCR. Thus, the produced expression library was judged to be sufficient for use in expression 
cloning. 

[0104] Fig. 4A and Fig. 4B show a flow diagram of the screening described below. In the first screening, 4000 inde- 

40 pendent clones were used in one pool, and 24 pools (corresponding to 96000 clones) were produced. The plasmids 
were packaged into retroviruses by transfecting each plasmid into BOSC23 cells. The resulting viruses derived from 
each pool were infected into NIH3T3 cells. Three days after infection, the cells were detached, and after staining with 
2D7 antibody, expression analysis was performed using FACS. As a result, compared to NIH3T3 cells infected with 
viruses derived from an empty vector (control), 2D7-positive cells were found in 3 of the 24 pools (pools 4, 13, and 21). 

45 [0105] Next, pools 4 and 13, which showed positive results in the first screening, were divided into four pools each 
comprising 1 000 independent clones, and a second screening was performed. As a result, a single clearly positive pool 
was found from each pool (Fig. 5A, pool 4-4, and pool 13-1). Pool 1 3-1 was further divided into 21 pools, each comprising 
1 60 independent clones, to perform a third screening. Two positive pools (Fig. 5B, 13-1 -1 1 and 13-1-21 ) were identified. 
Subsequently, pool 13-1 -1 1 was divided into eight pools, each comprising 20 clones, to perform a fourth screening, and 

so a positive pool (Fig. 5C, 13-1-11-5) was obtained. 

[0106] This pool was spread onto an LB plate, 64 colonies were picked one by one, and each of these were inoculated 
to one well of a 96-well plate. The eight clones in the vertical rows were taken as one pool to produce eight pools (1 to 
8), and the eight clones in the horizontal rows were taken as one pool to produce eight pools (A to H), and a fifth screening 
was performed. As a result, pools 3, 4, 6, and 8, and pools E, F, and G were positive, thus narrowing down the positive 

55 candidate clones to twelve clones (Fig. 6A). FACS was performed on these twelve clones, and ultimately four positive 
clones (3F, 4G, 6E, and 8G) were identified as a single clone recognized by the 2D7 antibody (Fig. 6B). 
[0107] As a result of reading the sequence of the insert portion of these clones, all four clones were found to be the 
full-length cDNA sequence of Human MHC class I HLA-A-6802. 



16 



EP 1 757 686 A1 



[0108] HLA-A is classified into several dozen types of haplotypes. As a result of this cloning, the A*6802 haplotype of 
HLA class I was identified as a 2D7 antigen, but since the 2D7 antibody recognizes a wide variety of cells, the haplotype 
of HLA class I in the RPMI8226 cells that were used as the gene source just happened to be A*6802, and the 2D7 
antibody was considered to be an antibody that recognizes any haplotype of HLA class I molecules. 

[Example 3] Examination of growth inhibitory effect 

[0109] Several types of leukemia cell lines (K562, Jurkat, and RPMI8226) were used to investigate whether the 2D7 
antibody has a cytocidal effect. The expression level of the 2D7 antigen in the three cell lines is: K562, weakly positive; 
Jurkat and RPMI8226, strongly positive. 

[0110] K562 and Jurkat cells were plated in the presence or absence of PHA and PMA, and 10 |j,g/ml of the 2D7 
antibody was added thereto. On observing the cells 24 hours later, weakly 2D7-positive K562 cells did not show obvious 
differences in their morphology due to the presence or absence of the 2D7 antibody, however, addition of 2D7 antibody 
resulted in significant cell aggregation in Jurkat cells strongly expressing 2D7 (Fig. 7B and Fig. 7C). However, growth 
inhibition due to addition of the 2D7 antibody was not observed (Fig. 7A). Growth inhibition due to 2D7 in Jurkat cells 
activated by PHA and PMA stimulation was also not observed. 

[0111] Unexpectedly, addition of 2D7 antibody did not have an obvious effect on the morphology and growth of the 
strongly 2D7-positive RPMI8226 cells (Fig. 7D). 

[0112] Next, it was examined whether cytocidal effects can be observed by adding anti-mouse IgG(Fc) antibody to 
2D7 antibody, to cross-link the antibodies. Anti-mouse IgG was added to Jurkat cells, in the presence or absence of 
2D7 antibody. The cells were cultured, and 48 hours later the cell nuclei were stained with Hoechsr.33258. Cells were 
observed for fragmentation of cell nuclei, which is characteristic of dead cells (Fig. 8). As a result, nuclear fragmentation 
was observed in Jurkat cells by further cross-linking 2D7 with an antibody, indicating that cell death was induced. 

[Example 4] Cloning of cDNA encoding the 2D7 antibody variable region, and the predicted diabody structure 

[0113] Primers for the constant regions of the heavy chain and light chain of mouse lgG2b were produced, and DNA 
encoding the 2D7 variable region was cloned by 5' RACE method. The nucleotide sequences of the obtained PCR 
products are shown in SEQ ID NO: 1 and 2. 

[0114] A single chain was then constructed based on these sequences. As shown in Fig. 9 and Fig. 10A, the 2D7 
single chain is composed of the leader sequence of the heavy chain, the variable region of the heavy chain, and then 
across from a 5mer linker (GGGGS), the variable region of a light chain, followed by a cDNA (SEQ ID NO: 3) encoding 
a Flag-tag. Dimerization of this single chain may cause the 2D7 diabody to form the structure shown in Fig. 10B. 

[Example 5] Analysis of the cytotoxic activity of the 2D7 diabody 

(i) Cytotoxic activity of the 2D7 diabody transiently expressed in COS7 

[0115] A 2D7 diabody expression vector was transfected into COS7 cells, and the culture supernatant was collected 
three days later. The culture supernatant and cell lysate were subjected to SDS-PAGE, and after performing Western 
blotting with an anti-Flag-tag antibody, a 2D7 single chain was found to be secreted in the culture supernatant (Fig. 1 0C). 
[0116] This culture supernatantwas added to Jurkat cells at a ratio of 50%. The percentage of dead cells was measured 
by staining the cells with PI and Annexin V a few days later. No significant change in the apoptosis marker was observed 
in Jurkat cells to which just the anti-BST-1 antibody and 2D7 antibody (5 |j,g/ml each) were added. Furthermore, no 
particular change could be observed when using the culture supernatant of COS7 transfected with the vector alone. On 
the other hand, cell death was clearly induced in Jurkat cells to which the culture supernatant of COS7 expressing 
2D7DB was added (Fig. 1 1 A and Fig. 1 1 B). 

[0117] Next, to investigate the HLA class I A-specific action of this 2D7DB, a similar experiment was performed using 
K562 cells, which are known to not express HLA class I A. As a result, 2D7DB had absolutely no influence on K562 
cells, although it showed cell death inducing activity against Jurkat cells (Fig. 12A and Fig. 12B). This strongly supports 
the idea that the cell death inducing activity of 2D7DB is an action targeting HLA class I A, which is its epitope. Furthermore, 
according to each data, the sensitivity of Jurkat cells towards 2D7DB was found to be slightly higher in cells stimulated 
by con A. 

[0118] Next, the action of 2D7DB on other myeloma cell lines was analyzed. RPMI8226, IL-KM3, U266, and ARH77 
were incubated with a culture supernatant into which the vector alone was transfected (control), or with the 2D7DB- 
expressing COS7 culture supernatant. Two days later these cultures were double stained with Annexin V and PI, and 
analyzed using a flow cytometer. As a result, incubation with 2D7DB was found to significantly induce cell death in all 
of the cells (Fig. 13A to Fig. 13D). 



17 



EP 1 757 686 A1 



(ii) Cytotoxic activity of purified 2D7DB 

[0119] The growth inhibitory effect of purified 2D7DB on each type of cell line (RPMI8226, ARH77, U266, and Jurkat) 
was analyzed. 2D7DB was added at 0, 0.5, 1 .0, and 2.0 |mg/ml, and the number of cells was counted three days later. 

5 As a result, 2D7DB was found to inhibit cell growth of these cells in a concentration-dependent manner (Fig. 14). 

[0120] Purified 2D7DB was then added, and 48 hours later, cells were stained with cell death markers, PI and Annexin 
V, and then analyzed. As in the results obtained when using 2D7DB transiently expressed in COS7, cell death was 
induced in Jurkat and ARH77 in a concentration-dependent manner, and K562 was not affected at all (Fig. 1 5A to Fig. 
15C). Furthermore, 48 hours after the addition of 2D7DB to U266 and IL-KM3, significant cell death inducing activity 

10 was confirmed (Fig. 1 6A and Fig. 1 6B). 

[0121] On the other hand, although the 2D7 antibody stained the adherent HeLa cells very well, 2D7DB had absolutely 
no influence under the same conditions (Fig. 15 D). This suggested that 2D7DB may act specifically on non-adherent 
cells, such as blood cells. 

[0122] Next, the time taken for 2D7DB to induce cell death was analyzed. 2 |j,g/ml of 2D7DB was added to ARH77 
15 and Jurkat cells, cells were collected 1 2, 24, and 38 hours later, and stained with a cell death marker. The results showed 
that cell death was already induced in all cells twelve hours later (Fig. 1 7A and Fig. 17B). Therefore, cell death induction 
was investigated at earlier times (three and six hours). Surprisingly, it was shown that 2D7DB induces cell death at least 
within three hours after its addition (Fig. 1 8A and Fig. 1 8B). These results strongly support the idea that 2D7DB has a 
very strong cell death-inducing activity. Since 2D7DB strongly induces cell death, sufficient drug efficacy can be expected 
20 even with a short half life in the blood. Furthermore, safety becomes a concern if the whole antibody has strong cell 
death-inducing activity, considering the length of the half life in the blood; however, producing a diabody can overcome 
such problems. 

[0123] Next, analyses were performed to determine whether cell death due to 2D7DB is induced through caspase 
activation, that is, whether it is apoptosis. As shown in Fig. 19 and Fig. 20, significant apoptosis was induced when 

25 ARH77 and Jurkat cells were treated with the apoptosis-inducing agent Actinomycin D, and then stained 1 6 hours later 
with Annexin V and PI. After pre-treating cells under these conditions with caspase inhibitor Z-VAD-FMK for 2.5 hours, 
apoptosis due to Actinomycin D was suppressed. However, cell death induced by 2D7DB was not inhibited at all by 
pretreatment with Z-VAD-FMK. These results show that 2D7DB induces cell death by a mechanism different from the 
ordinary caspase-mediated apoptosis mechanism. 

30 [0124] To confirm this, fragmentation of chromatin DNA, known to be the most characteristic biochemical change 
accompanying apoptosis, was also analyzed. 

[0125] ARH77 and Jurkat cells were treated with 2D7DB (2 |j-g/ml) or Actinomycin D, and DNAs were collected from 
the cells 24 hours later and subjected to electrophoresis (Fig. 21). As a result, DNA fragmentation characteristic of 
apoptosis was induced in all cells treated with Actinomycin D, which is an apoptosis-inducing agent. On the other hand, 
35 DNA fragmentation was not observed at all in 2D7DB-treated cells, even though the concentration of added 2D7DB was 
absolutely sufficient to induce cell death. These results also strongly support the idea that cell death due to 2D7DB is 
an unknown type of cell death, unaccompanied by the characteristics of apoptosis. 

[0126] From the above-mentioned results, cell death due to 2D7DB was found to be caused by a pathway different 
from previously known cell death induction mechanisms. Therefore, further analysis was performed to elucidate the 
40 mechanism of cell death induction by 2D7DB. From the experiments described above, when 2D7DB was reacted with 
the cells, the cell membranes were often observed to be destroyed under the microscope. Therefore, 2D7DB was 
presumed to have some sort of influence on the actin skeleton. To examine this possibility, the cells were treated with 
an actin polymerization inhibitor (cytochalasin D), and then the influence of 2D7DB on cell death induction activity was 
analyzed. 

45 [0127] Cytochalasin D (20 |j,g/ml) or ethanol alone (control) was added to ARH77 cells, and 1 hour later, 2D7DB was 
added at various concentrations. After a 4-hour incubation from the 2D7DB addition, cells were collected, PI staining 
was performed and the percentage of dead cells was measured (Fig. 22). As a result, pretreatment of cells with cyto- 
chalasin D was found to cause loss of sensitivity towards 2D7DB. These results suggested that 2D7DB causes some 
kind of effect on the cytoskeletal system such as actin to induce cell death by binding to HLA-class IA, which is the target 

so molecule. 

[0128] Therefore, cells treated with 2D7DB were stained by the actin antibody, and the dynamic change of the cy- 
toskeletal system due to 2D7DB addition was analyzed visually. ARH77 cells were treated with 2D7DB, and 1 5 minutes 
later, the cells were immobilized with methanol, and the state of actin (red) in the cells was investigated by immunostaining 
(Fig. 23). As a result, compared to the image from those not treated with 2D7DB, significant destruction of the actin 
55 skeleton in the cell due to 2D7DB was observed. 

[0129] The above-mentioned results strongly suggested that cell death due to 2D7DB may be caused by destruction 
of the actin skeleton in cells by 2D7DB bound to HLA class IA. This is a completely new type of cell death induction 
mechanism that has not been reported to date. 



18 



EP 1 757 686 A1 



[Example 6] Drug efficacy test for 2D7 diabody using human myeloma animal model 

(1) Production of mouse model for human myeloma 

5 [0130] A mouse model of human myeloma was produced as follows. ARH 77 cells were prepared to reach 2. 5x 10 7 
cells/ml in RPMI1 640 medium (GIBCO BRL) supplemented with 1 0% fetal calf serum (GIBCO BRL), and then 200 |jlL 
of the above-mentioned ARH77 cell suspension (5x 10 6 cells/mouse) was injected to SCID mice (male, 6 weeks old, 
Clea Japan) pretreated the day before with intraperitoneal administration of 0.2 mg of anti-asialo GM1 antibody (Wako 
Pure Chemicals) from the tail vein. 

10 

(2) Preparation of the antibody to be administered 

[0131] On the day of administration, a 2D7 diabody was prepared at 0.8 mg/ml using filter-sterilized PBS(-), and this 
was used as the administration sample. 

15 

(3) Antibody administration 

[0132] To the mouse model of human myeloma produced in (1), the administration sample prepared in (2) was ad- 
ministered through the tail vein at 1 0 ml/kg twice a day for 3 days from the first day after engraftment of ARH77 cells. 
20 As a negative control (vehicle), filter-sterilized PBS(-) was administered similarly at 10 ml/kg through the tail vein twice 
a day for 3 days. The antibody-administered group had 7 animals per group, and the vehicle-administered group had 8 
animals per group. 

(4) Human IgG assay of mouse serum 

25 

[0133] The quantity of human IgG produced by human myeloma cells in the mouse serum was determined by ELISA 
described below. 1 00 |j,Lof goat anti-human IgG antibody (BIOSOURCE) diluted to 1 |j,g/ml with 0.1 % bicarbonate buffer 
(pH9.6) was placed into a 96-well plate (Nunc), this was incubated at 4°C overnight, and the antibody was immobilized. 
After blocking, mouse serum diluted in a stepwise manner, or as a standard, 1 00 (j. L of human IgG (Cappel) was added, 
30 and this was incubated at room temperature for 1 hour. After washing, 100 |jlL of a5000-fold diluted alkaline phosphatase- 
labeled anti-human IgG antibody (BIOSOURCE) was added, and this was incubated at room temperature for 1 hour. 
After washing, substrate solution was added, and after incubation, absorbance at 405 nm was measured using MICRO- 
PLATE READER Model 3550 (BioRad), and the concentration of human IgG in mouse serum was calculated from the 
calibration curve obtained from the absorbance of the standard human IgG sample. 

35 

(5) Evaluation of anti-tumor effect 

[0134] The anti-tumor effect of the 2D7 diabody on a human myeloma mouse model was evaluated using the change 
in the amount of human IgG (M protein) produced by the myeloma cells in mouse serum, and by the survival time. 

40 Regarding the change in human IgG level in mouse serum, serum was collected on the 24th day after transplanting the 
ARH77 cells, and the human IgG level was measured by the ELISA described above in (4). As a result, the level of 
human IgG (M protein) in the serum had increased in the vehicle-administered group to approximately 74 |j,g/ml. In 
contrast, the level in the 2D7 diabody-administered group was significantly lower than in the control group (P<0.005, 
unpaired t-test), and 2D7 diabody was shown to very strongly suppress the growth of ARH77 cells (Fig. 24). With regards 

45 to survival time, as shown in Fig. 25, the 2D7 diabody-administered group showed a significant increase in survival time 
compared to the vehicle-administered group. 

[0135] Accordingly, the 2D7 diabody was shown to have an antitumor effect on the mouse model of human myeloma. 
The antitumor effect of the2D7 diabodies of this invention maybe based on the cell death-inducing action of this antibody. 

so [Example 7] Analysis of the action of 2D7DB on PBMC 

[0136] The action of 2D7DB on human peripheral blood mononuclear cells (PBMCs) was analyzed. PBMCs were 
purified from the peripheral blood of a healthy adult volunteer by density gradient centrifugation. The PBMCs were plated 
at 5x 1 0 5 cells/1 ml/well onto a 24-well plate, in the presence or absence of a mitogen. Phytohemagglutinin M (PHA-M, 
55 Roche Diagnostics, final concentration: 10 |j,g/ml), concanavalin A (ConA, Wako, final concentration: 10 |j,g/ml), and 
SAC (Pansorbin Cells, Calbiochem, final concentration: 0.01 %) were used as mitogens. Cells were cultured in a 5% 
C0 2 incubator at 37°C for three days. 24 or 3 hours before culture was complete, 2D7DB was added to yield a final 
concentration of 2 |j,g/ml. After culture was complete, the cells were double stained with Annexin V and PI (Annexin V- 



19 



EP 1 757 686 A1 



FITC Apoptosis Detection Kit I, Pharmingen), and then analyzed using a flow cytometer (EPICS XL, Coulter). As a 
positive control, ARH77 at 2.5x 1 0 5 cells/ml/well was cultured for 24 hours in the absence of a mitogen, and was reacted 
with 2D7DB, as for PBMC. 

[0137] In the case of PBMC, the percentages of dead cells that were both Annexin V and Pl-positive were 29%, 23%, 
5 and 25% in the absence of mitogens (in order: no addition, 3-hour addition, and 24-hour addition of 2D7DB; continued 
below); 20%, 45%, and 42% in the presence of PHA-M; 22%, 30%, and 34% in the presence of ConA; and 31%, 38%, 
and 40% in the presence of SAC (Figs. 26A to 26D). In the case of ARH77, the percentages were 16%, 56%, and 58% 
(Fig. 26E). These results showed that 2D7DB has hardly any effect on unstimulated PBMC, but induces cell death in a 
short time with mitogen-activated PBMC. 

10 

Industrial Applicability 

[0138] This invention provides minibodies with high specific activities. By using these minibodies, adequate drug 
efficacy can be expected even with a short half-life. The minibodies of the present invention are further expected to be 
15 able to improve drug efficacy and to lower toxicity. In addition, since overall cost is reduced, including reduction of clinical 
dose and production cost, economical problems of concern in the development of antibody pharmaceuticals are also 
expected to improve. 



20 



EP 1 757 686 A1 

SEQUENCE LISTING 

<110> CHUGAI SEIYAKU KABUSHIKI KAISHA 
OZAKI Shuji 
ABE Masahiro 

<120> Cell Death Inducer 

<130> C1-A0404P 

<160> 18 

<170> Patentln version 3. 1 

<210> 1 

<211> 547 

<212> DNA 

<213> Mus musculus 

<400> 1 

tacgactcac tatagggcaa gcagtggtat caacgcagag tacgcgggga atctatgatc 
agtgtcctct ctacacagtc cctgacgaca ctgactccaa ccatgcgatg gagctggatc 
tttctcttcc tcctgtcaat aactgcaggt gtccattgcc aggtccagtt gcagcagtct 



EP 1 757 686 A1 



ggacctgagc tggtgaagcc tggggcttca gtgaagatgt cttgtaaggc ttctggctac 240 



accttcacag actactttat acactgggtg aaacagaggc ctggacaggg acttgaatgg 300 



attggatgga tttttcctgg agatgatact actgattaca atgagaagtt caggggcaag 360 



accacactga ctgcagacaa atcctccagc acagcctaca ttttgctcag cagcctgacc 420 

15 



tctgaggact ctgcgatgta tttctgtgta aggagtgacg actttgacta ctggggccag 480 

20 

ggcaccactc tcacagtctc ctcagccaaa acaacacccc catcagtcta tccactggcc 540 

25 

cctgctg 547 

30 

<210> 2 
<211> 535 

35 

<212> DNA 

<213> Mus musculus 

40 

<400> 2 

ctaatacgac tcactatagg gcaagcagtg gtatcaacgc agagtacgcg gggactwatg 60 

45 

agaatagcag taattagcta gggaccaaaa ttcaaagaoa aaatgcattt tcaagtgcag 120 

50 ■ ., . 

attttcagct tcctgctaat cagtgcctca gtcatcatgt ccagaggaca aattgttctc 180 



22 



EP 1 757 686 A1 

acccagtcgc cagcaatcat gtctgcatct ccaggggaga aggtcaccat aacctgcagt 

gccagctcaa gtgtaagtta catgcactgg ttccagcaga agccaggcac ttttcccaaa 

ctctggattt atagcacatc caacctggct tctggagtcc ctactcgctt cagtggcagt 

ggatctggga cctcttactc tctcacaatc agccgaatgg aggctgaaga tgctgccact 

tattactgcc agcaaaggac gagttatcca cccacgttcg gctcggggac aaagttggag 

ataaaacggg ctgatgctgc accaactgta tccatcttcc caccatccag tgagc 

<210> 3 

<211> 789 

<212> DNA 

<213> Artificial 

<220> 

<223> an artificially synthesized DNA sequence 
<220> 

<221> CDS 

<222> (14). . (775) 

<223> : 



23 



EP 1 757 686 A1 



<400> 3 

cctgaattcc acc atg cga tgg age tgg ate ttt etc ttc etc ctg tea 

Met Arg Trp Ser Trp lie Phe Leu Phe Leu Leu Ser 
1 5 10 

ata act gca ggt gtc cat tgc cag gtc cag ttg cag cag tct gga cct 
He Thr Ala Gly Val His Cys Gin Val Gin Leu Gin Gin Ser Gly Pro 
15 20 25 

gag ctg gtg aag cct ggg get tea gtg aag atg tct tgt aag get tct 
Glu Leu Val Lys Pro Gly Ala Ser Val Lys Met Ser Cys Lys Ala Ser 
30 35 40 

ggc tac acc ttc aca gac tac ttt ata cac tgg gtg aaa cag agg cct 
Gly Tyr Thr Phe Thr Asp Tyr Phe He His Trp Val Lys Gin Arg Pro 
45 50 55 60 

gga cag gga ctt gaa tgg att gga tgg att ttt cct gga gat gat act 
Gly Gin Gly Leu Glu Trp He Gly Trp He Phe Pro Gly Asp Asp Thr 
65 70 75 

act gat tac aat gag aag ttc agg ggc aag acc aca ctg act gca gac 
Thr Asp Tyr Asn Glu Lys Phe Arg Gly Lys Thr Thr Leu Thr Ala Asp 
80 85 90 



24 



EP 1 757 686 A1 



aaa tec tec age aca gec tac att ttg etc age age ctg ace tct gag 
Lys Ser Ser Ser Thr Ala Tyr lie Leu Leu Ser Ser Leu Thr Ser Glu 
95 100 105 



gac tct gcg atg tat ttc tgt gta agg agt gac gac ttt gac tac tgg 
Asp Ser Ala Met Tyr Phe Cys Val Arg Ser Asp Asp Phe Asp Tyr Trp 
110 115 120 



ggc cag ggc acc act etc aca gtc tec tea ggt gga ggc ggt age caa 

Gly Gin Gly Thr Thr Leu Thr Val Ser Ser Gly Gly Gly Gly Ser Gin 

125 130 135 140 

att gtt etc acc cag teg cca gca ate atg tct gca tct cca ggg gag 

He Val Leu Thr Gin Ser Pro Ala He Met Ser Ala Ser Pro Gly Glu 

145 150 155 



aag gtc acc ata acc tgc agt gec age tea agt gta agt tac atg cac 
Lys Val Thr He Thr Cys Ser Ala Ser Ser Ser Val Ser Tyr Met His 
160 165 170 



tgg ttc cag cag 
Trp Phe Gin Gin 
175 

aca tec aac ctg 
Thr Ser Asn Leu 



aag cca ggc act 
Lys Pro Gly Thr 
180 

get tct gga gtc 
Ala Ser Gly Val 



ttt ccc aaa etc 
Phe Pro Lys Leu 

cct act cgc ttc 
Pro Thr Arg Phe 



tgg att tat age 
Trp lie Tyr Ser 
185 

agt ggc agt gga 
Ser Gly Ser Gly 



25 



EP 1 757 686 A1 



190 195 200 

tct ggg acc tct tac tct etc aca ate age cga atg gag get gaa gat 
Ser Gly Thr Ser Tyr Ser Leu Thr He Ser Arg Met Glu Ala Glu Asp 
205 210 215 220 

get gec act tat tac tgc cag caa agg acg agt tat cca ccc acg ttc 
Ala Ala Thr Tyr Tyr Cys Gin Gin Arg Thr Ser Tyr Pro Pro Thr Phe 
225 230 235 

ggc teg ggg aca aag ttg gag ata aaa gac tac aag gat gac gac gat 
Gly Ser Gly Thr Lys Leu Glu He Lys Asp Tyr Lys Asp Asp Asp Asp 
240 245 250 



aag tga taagcggccg caat 
Lys 



<210> 4 

<211> 253 

<212> PRT 

<213> Artificial 

<220> 

<223> an artificially synthesized peptide sequence 



26 



EP 1 757 686 A1 



<400> 4 

Met Arg Trp Ser Trp He Phe Leu Phe Leu Leu Ser He Thr Ala Gly 
1 5 10 15 

Val His Cys Gin Val Gin Leu Gin Gin Ser Gly Pro Glu Leu Val Lys 
20 25 30 

Pro Gly Ala Ser Val Lys Met Ser Cys Lys Ala Ser Gly Tyr Thr Phe 
35 40 45 

Thr Asp Tyr Phe He His Trp Val Lys Gin Arg Pro Gly Gin Gly Leu 
50 55 60 

Glu Trp He Gly Trp He Phe Pro Gly Asp Asp Thr Thr Asp Tyr Asn 
65 70 75 80 



Glu Lys Phe Arg Gly Lys Thr Thr Leu Thr Ala Asp Lys Ser Ser Ser 
85 90 95 



Thr Ala Tyr He Leu Leu Ser Ser Leu Thr Ser Glu Asp Ser Ala Met 
100 105 110 



Tyr Phe Cys Val Arg Ser Asp Asp Phe Asp Tyr Trp Gly Gin Gly Thr 
115 120 125 



Thr Leu Thr Val Ser Ser Gly Gly Gly Gly Ser Gin He Val Leu Thr 



27 



EP 1 757 686 A1 

130 135 140 

Gin Ser Pro Ala He Met Ser Ala Ser Pro Gly Glu Lys Val Thr He 
145 150 155 160 

Thr Cys Ser Ala Ser Ser Ser Val Ser Tyr Met His Trp Phe Gin Gin 
165 170 175 

Lys Pro Gly Thr Phe Pro Lys Leu Trp He Tyr Ser Thr Ser Asn Leu 
180 185 190 

Ala Ser Gly Val Pro Thr Arg Phe Ser Gly Ser Gly Ser Gly Thr Ser 
195 200 205 

Tyr Ser Leu Thr He Ser Arg Met Glu Ala Glu Asp Ala Ala Thr Tyr 
210 215 220 

Tyr Cys Gin Gin Arg Thr Ser Tyr Pro Pro Thr Phe Gly Ser Gly Thr 
225 230 235 240 

Lys Leu Glu He Lys Asp Tyr Lys Asp Asp Asp Asp Lys 
245 250 

<210> 5 
<211> -29 



28 



EP 1 757 686 A1 



<212> DNA 

<213> Artificial 

<220> 

<223> an artificially synthesized adapter sequence 



<400> 5 

15 

aattcccagc acagtggtag ataagtaag 29 



20 



35 



<210> 6 

<211> 29 

<212> DNA 

<213> Artificial 

<220> 

<223> an artificially synthesized adapter sequence 

<400> 6 

tcgacttact tatctaccac tgtgctggg 29 



<210> 7 

<211> 24 

<212> DNA 

<213> Artificial 



29 



EP 1 757 686 A1 



<220> 

<223> an artificially synthesized primer sequence 
<400> 7 

caggggccag tggatagact gatg 

<210> 8 

<211> 23 

<212> DNA 

<213> Artificial 

<220> 

<223> an artificially synthesized primer sequence 
<400> 8 

gctcactgga tggtgggaag atg 



<210> 9 

<211> 35 

<212> DNA 

<213> Artificial 

<220> 



30 



EP 1 757 686 A1 

<223> an artificially synthesized primer sequence 

<400> 9 

cctgaattcc accatgcgat ggagctggat ctttc 

<210> 10 

<211> 47 

<212> DNA 

<213> Artificial 

<220> 

<223> an artificially synthesized primer sequence 
<400> 10 

aatttggcta ccgcctccac ctgaggagac tgtgagagtg gtgccct 



<210> 11 

<211> 47 

<212> DNA 

<213> Artificial 

<220> 

<223> an artificially synthesized primer sequence 



31 



EP 1 757 686 A1 



<400> 11 

tcctcaggtg gaggcggtag ccaaattgtt ctcacccagt cgccagc 

<210> 12 

<211> 68 

<212> DNA 

<213> Artificial 

<220> 

<223> an artificially synthesized primer sequence 
<400> 12 

attgcggccg cttatcactt atcgtcgtca tccttgtagt cttttatctc caactttgtc 
cccgagcc 



<210> 13 

<211> 5 

<212> PRT 

<213> Mus musculus 

<400> 13 

Asp Tyr Phe lie His 
1 5 



32 



EP 1 757 686 A1 



<210> 14 

<211> 17 

10 <212> PRT 

<213> Mus musculus 

15 

<400> 14 

Trp lie Phe Pro Gly Asp Asp Thr Thr. Asp Tyr Asn Glu Lys Phe Arg 
20 1 5 10 15 

Gly 



35 



40 



45 



50 



<210> 15 

<211> 6 

<212> PRT 

<213> Mus musculus 

<400> 15 

Ser Asp Asp Phe Asp Tyr 
1 5 



<210> 16 
<211> 10 



33 



EP 1 757 686 A1 



<212> PRT 

<213> Mus musculus 



35 



45 



50 



55 



<400> 16 

Ser Ala Ser Ser Ser Val Ser Tyr Met His 
1 5 10 



15 

<210> 17 
20 <211> 7 



<212> PRT 

<213> Mus musculus 

<400> 17 

Ser Thr Ser Asn Leu Ala Ser 
1 5 



<210> 18 

<211> 9 

<212> PRT 

<213> Mus musculus 

<400> 18 

Gin Gin Arg Thr Ser Tyr Pro Pro Thr 

, . 1 5 

Claims 

1. A minibody, which comprises a heavy chain variable region comprising CDRs 1 , 2, and 3 which consist of the amino 



34 



EP 1 757 686 A1 

acid sequences of SEQ ID NOs: 13, 14, and 15. 

2. A minibody which is functionally equivalent to the minibody of claim 1 , and which comprises heavy chain CDRs 
consisting of amino acid sequences with one or more amino acid substitutions, deletions, insertions, and/or additions 

5 in the amino acid sequences of the heavy chain CDRs of the minibody of claim 1 . 

3. A minibody, which comprises a light chain variable region comprising CDRs 1 , 2, and 3 which consist of the amino 
acid sequences of SEQ ID NOs: 16, 17, and 1 8. 

10 4. A minibody which is functionally equivalent to the minibody of claim 3, and which comprises light chain CDRs 
consisting of amino acid sequences with one or more amino acid substitutions, deletions, insertions, and/or additions 
in the amino acid sequences of the light chain CDRs of the minibody of claim 3. 

5. A minibody, which comprises a heavy chain variable region comprising CDRs 1 , 2, and 3 which consist of the amino 
15 acid sequences of SEQ ID NOs: 13, 14, and 15, and a light chain variable region comprising CDRs 1 , 2, and 3 which 

consist of the amino acid sequences of SEQ ID NOs: 16, 17, and 18. 

6. A minibody which is functionally equivalent to the minibody of claim 5, and which comprises CDRs consisting of 
amino acid sequences with one or more amino acid substitutions, deletions, insertions, and/or additions in the amino 

20 acid sequences of the CDRs of the minibody of claim 5. 

7. The minibody of any one of claims 1 to 6, which is a diabody. 

8. A cell death-inducing agent comprising the minibody of any one of claims 1 to 7 as an active ingredient. 

25 

9. The cell death-inducing agent of claim 8, which induces cell death of a B-cell or T-cell. 

10. The cell death-inducing agent of claim 9, wherein the B-cell or T-cell is an activated B-cell or an activated T-cell. 
30 11. A cell growth-suppressing agent comprising the minibody of any one of claims 1 to 7 as an active ingredient. 

12. An anti-tumor agent comprising the minibody of any one of claims 1 to 7 as an active ingredient. 

13. The anti-tumor agent of claim 1 2, wherein the tumor is blood tumor. 



35 



45 



14. An agent for treating an autoimmune disease, wherein the agent comprises the minibody of any one of claims 1 to 
7 as an active ingredient. 



50 



35 



EP 1 757 686 A1 



FIG. 1 



5 ' -AATTCCCAGCACAGTGG TAGATAA G TAA G ( SEQ ID NO: 5 ) 

GGGT CGTGTCACC AT CTATT CAT T CAGCT - 5 ' ( SEQ ID NO: 6 ) 



36 



EP 1 757 686 A1 



FIG. 2A 





Q 

CM 



O 
Q 




o 



84 63 42 21 



146 109 73 36 0 



Q 

CM 

■ 

CD 
CM 
CM 
CO 



Q_ 




C3 



116 87 58 29 0 




85 63 42 21 



37 



EP 1 757 686 A1 




38 



EP 1 757 686 A1 



FIG. 3 



NIH3T3 RPMI8226 U266 




39 



EP 1 757 686 A1 




40 



EP 1 757 686 A1 



FIG. 5A 




41 



EP 1 757 686 A1 



FIG. 5B 




-\ — i i 1 una' i i n i i i iT "i 1 1 mm 
-1 

FITC 



TTTTT 

1000 



in in ii f t ii iiiir TT i n i in — r rumi 

-1 RTC 1000 



C_3 




13-1-1 



i 1 1 1 urn i 1 1 mm 



FITC 



1000 



CJ5 




13-1-2 



i 1 1 i imi — i n inn 



1000 




13-1-3 



-1 



i i null i TTTTr m — i 1 1 hub — i i t i m i 



FITC 



1000 




i i ii inn " t i TTiiiti I i i huh — rm™ 

1 FITC 1000 




13-1-11 



nun i 1 1 1 inn t i iir m — niinir 

-1 RTC 10 oo 




13-1-12 



tttti i mrnn i i 



FITC 



1 1 Minn 
1000 



42 



EP 1 757 686 A1 



FIG. 5C 




CO 



-1 



10 100 1000 
FITC 




13-1-11 



10 100 1000 
FITC 




13-1-11-1 



i i 1 1 inn — i ri i mi 
10 100 1000 
FITC 




■1 1 



10 100 1000 
FITC 




i i i mm 1 Ti i i ii i T~ 1 "f " I I llllll 1 I l l J ill 

■ 1 1 10 100 1000 

FITC 




13-1-11-4 



\ i i i nun i iii iimi i 'i ' i i mi l — i i 1 1 mi 



-1 



1 10 100 1000 

FITC 




13-1-11-5 



"• ■ r i T i n hi 



* 

— I I i i i ii i i 1 — i i mm I — l llllll 

10 100 1000 
FITC 




10 100 1000 
FITC 



cj j mm 



13-1-11-7 



\ 1 — i i i i nn 1 I I Trim f n 



TTTTTl 1 I llllll 




13-1-11-8 



rffWnr^i r 1 1 : iTii : ' i 1 1 1 mi — i i 1 1 rm 



1 10 100 1000 -1 

FITC 



10 100 1000 
FITC 



43 



EP 1 757 686 A1 




44 



EP 1 757 686 A1 



FIG. 6B 




1 10 100 1000 
FITC 



1 10 100 1000 
FITC 




1 10 100 1000 
FITC 



1 10 100 1000 
FITC 



45 



EP 1 757 686 A1 



FIG. 7 A 



2.5 



E 1.5 

C-3 



QQ 1 



0.5 




□ PMA(-)2D7C-) 

■ PMA(-)2D7(+) 

■ PMA(+)2D7(-) 
H PMA(+)2D7(+) 



K562 



Jurkat RPMI8226 



FIG. 7C 

PHA/PMA(-)2D7(-) PHA/PMA(-)2D7(+) 



FIG 7B 

PHA/PMA(-)2D7(-) PHA/PMA(-)2D7(+) 



PHA/PMA(+)2D7(-) PHA/PMA(+)2D7(+) PHA/PMA(+)2D7(-) PHA/PMA(+)2D7(+) 







FIG 7D 2D70 




46 



EP 1 757 686 A1 



FIG. 8 




47 



EP 1 757 686 A1 



FIG. 9 



cd 


CD 






cn CD 


CD 


C3 


CD 






CD 


Eh 




CD 




CO 


CD 


EH 






CD 


CD 










o 








CD CD 




CD 


<! 


CO 




CD 


CJ 




CD 


CJ 




CD 


CD 


> 




CD 


CD 










i-H 














3 






^ 


< 


Eh 


LO 


CD 




ID 


<: 






[ — 




CC 










E— i 






o 


o 




CJ 








CJ 






Eh 


CJ 




CD 








S3 












o 






<; 






E-h 


CO 






CJ 






CJ 






CD 


CD 


















CJ 


Oh 




cd 






O 








< 


E-h 




CJ 






E-h 








CJ 


o 
















cd 


CD 




CJ 








CJ 








E-h 




CJ 








CD 












CD 






E—i 






E-h 


CO 






CD 






< 






Eh 


CO 






<l 












CD 


CD 




CJ 














CD 


CD 




E-h 






E-h 








CJ 


Of 








£h 






CJ 


On 












CD 






< 


t — i 




CJ 








CJ 










o 


C J 






CD CD 




CD 








CD 


e£ 




CD 


CJ 




CD 


CD 




CD 


ID 




o 






o\ 


Eh 


CO 




CTt CD 






o 






CTl 


o 


o 




CJ 




CTt 


CD 






CTi 


Eh 












CD 






* — 1 fc£ 


DC 










ro 


o 






^ 


Eh 


LO 


E-H 






vD 


CJ 




c 


rcS 












CD 






CD 


Q 






CD 






CJ 






CJ 








f< 






n3 






CJ 


o 




<c 






rt! 








CD 


CD 




E-h 






CJ 








Eh 


>H 




O 






CD 






3 


o 




O 








CD 






CD 


;> 












Eh 






cn 


















CD 


<; 






CD 






CD 






^ 








s, 












CJ 


o 










E-h 








E-h 












CJ 








Eh 


>-l 




(j) 






cd 












CJ 








CJ 








i< 




CJ 








Eh 






CTi 






E— i 






CD 






f=t| 


E-H 






kC, 






CD 






E-h 


CO 






CJ 






Cn 




cd 


E-h 






CD E-t 






CD 






CD 


E — i 


>H 


CD 


f=£| 




CD 


i=C 






CD 


rtl 


E-i 




(J) 




CO 


CD 






co CD 




OO 


E-h 






oo 


CJ 




CO 


CD 


CJ 


OO 


CJ 






CO 


CJ 




CO 


Cn 












i— i CD 




(X 


CJ 


,_q 




ro 


<! 




-^T 


CD 




LO 


<! 


Eh 




LO 


CJ 




[ — ■ 


co 






cj 


o 




CD 






<! 








CD 


Q 




CD 






3 








CD 







n3 






CJ 






E-h 






CJ 








Eh 






CD 


CD 




CD 








Eh 






-M 






Eh 






o 








Eh 






E-h 






< 








CO 






CJ 






03 






CD 


> 










CJ 








E-h 


Du 




CJ 






E-H 








CD 






Cn 






CD 






O 






CJ 








CJ 






CJ 


Dj 












Eh 












3. 






<! 






<C 


Eh 






<C 






Eh 






E-h 


>H 












Cn 






CJ 


o 




E-h 






CD 








CD 


Q 




CJ 






E-H 








CD 


_ 








o 








CD fc£ 


HH, 


CD 








CD 


CJ 




CD 


Eh 


CO 


CD 


Eh 






CD 






d> 


n3 




r~ 


cd 






I — E-h 






^ 


JiS 






<! 






f=C 






<! 


I — | 






^ 






4_0 






E— i 


cj 




i— 1 E-h 






CJ 






ro 


CD 


Q 




CJ 




LO 


CD 






CO 


CD 




V 


ro 






E-h 






E-h 


Du 




CD 








E-h 






CD 






CD 








Eh 






CnQ i 




i< 






cj 






CD 


CD 






CD 






E-h 






Eh 








CJ 






O 






CJ 


ac 




<; 






CD 










CO 




CJ 






CJ 








CD 






fO 






cj 






E-H 


>H 




CD 








3 






E-h 


CO 




Eh 








CD 






cn Q ! 




Eh 






cj 






<C 


CC 






CD 






CD 






CJ 








<C 






u 






cd 


> 




<; 






CJ 








<< 


Pi 




Eh 














CD 


f-Lj 




(0 






E-H 






cd 


Q 




E-h 








< 






<C 














CD 






Cn Q 


cd 


CD 






o f=d 




CD 


E-h 


Cm 




CD 


Eh 




CD 


CJ 




CD 








o 


Eh 




Cl^ 


4-J 




lo 


CD 


cd 




lo CJ 






CD 






to 


CD 


>. 


CD 


Eh 




LO 


CJ 






CO 


st! 






03 






< 






* — ! 


EH 


•Cn] 








ro 


Eh 




-^r 




i — r 


LO 


CJ 






LO 






r 


Cn Q i 




(_) 






CJ 






^ 








CD 












CJ 


Q-i 






CD 






Cn 


ICO 




CD 


<! 




E-H 






CD 








E-H 


CJ 




CJ 






Eh 








CJ 






f0 




E-h 






E-h 


Cu 




<c 




l3 




CJ 






CD 


< 




Eh 




CD 




CJ 






rO 






CJ 






CJ 






CD 


&] 




Eh 






3, 






Eh 


Cjh 






CD 






O 








E-H 




CJ 






E-h 








E-h 


Du 




CJ 






Eh 




LU 

or: 






L/J 




ro 










<; 


E-h 








1 1 l 




E-h 






CJ 


Oh 




CJ 






o 






4-J 






E-H 






cj 








:z 


1 




<£ 






CD 






ct, 


Eh 


1 1 i 




Eh 






(J 




CD 




1 — 1 


LO 

CJ5 


o <C 




C3 


CJ 




CO 


CD 


E-h 


>H 


CD 


CJ 




CD 


CJ 




an 


o 


^ 


1 — 1 




rO 




LO 


^ 




LO E-H 


>H 


LT) 








LO 


CD 




LO 


E-h 


CO 


LO 


CD 




LO 






L-O 


cn Q i 




CJ 




U_l 


i— I CJ 




Cs] 


Eh 


>H 




CO 


E-h 






CD 




LO 


CD 


CD 




LO 


CJ 




r 








E-h 


CO 


— -H 


cd 






E-h 








<c 












<=c 




or 




eH 


b — 1 










CD 




LU 


CD 


CD 




<l 








CD 






CJ 


C* 


CJ 








CJ 








^ i 




E-h 




E-h 






CD 


Q 


- — 




CJ 






CJ 






CJ 


Oh 






Eh 












CJ 


hh 1 




cj 






E-h 








CD 






CJ 






CD 




- ^ 




o 


i — 1 










O 




t-L-J 


E-h 


CO 




CJ 




co> 




E-H 






< 


fcH 




< 








EH 












E-h 






E-h 






Pt; 


Eh 




CJ 






CJ 








sc; 


nz 
i 




CJ 






CD 






CJ 




Ll_l 


CJ 














f^H 


co 




Eh 






3 






Eh 


L/J 




< 




CD 


c_> 






CD CD 


<! 


CD 


CJ 






CD 


CJ 




CD 


CJ 


HH 


CD 






~r- 


CD 


CJ 




o 


CD 


w j 




E-h 






CD 






f< 


Eh 


i ■ i 










Eh 






3 


o 






< 






CD 






E-i 







» — i f£& 




Osl 


E-h 




n= 


ro 


3 


Q 




Eh 




LO 


CD 






LO 


Eh 


►> 1 


r 


Eh 






CJ 












f< 








CD 






CD 


> 




< 




1 




Eh 






Eh 


i-H- ! 




Ei 






Eh 






CD 


Q 












C ' 






CJ 


o 






CJ 






CD 






cj 


i_q 


1 


CD 






E-h 








o 


Cd 




E-h 






CJ 








EH 


C/j 










E-h 






E-h 


CJ 




<< 








Eh 








i — 1 




Eh 








CJ 












&H 






E-H 






CD 


Q 






CJ 












Eh 


ClH 






CJ 












E-h 






O 






^ 








Eh 


co 










CD 








< 


b — ' 




O 






CJ 






Fh 


CO 




CD 








CJ 






CJ 


oc 


CD 








3 






< 


EH ; 


cd 


E-H 






CD CD 




CD 


CD 


CD 




CD 


CJ 




CD 


CJ 




CD 


Eh 






CD 


CD 




r . 

~r 


CD 




co 


< 


I 1 




CO E-h 




m 


F-h 






ro 


<! 


Eh 


ro 


Cn 




ro 


CJ 






ro 


CD 


*l_J 


CM 


CD 






CD 






i — i <C 




CS] 


CJ 






ro 


CD 






ro 


co 


LO 


< 






LO 


Eh 






CD 


CD| 




CD 






CD 






CJ 


Dh 






Eh 












CJ 


ca 






CJ 






CD 






E-h 






<c 






E-h 








CJ 


i-^l 




Cn 






CD 








Eh 


CO 




O 






CJ 








5seJ 




E-h 








CJ 






Cn CD 




Eh 








< 






Eh 


CO | 




CD 






CD 






E-h 


tu 






CD 






CJ 






< 








CD 






CJ 






< 


co 




EH 






E-h 








< 


CO 




Cn 






CJ 








CD 






CD 






CD 






CD 


> 




E-h 








CJ 






Cn CD 




«i 








Eh 






CD 


CD i 




cd 












r< 


1 — l 






CD 






ro 






Eh 


>H 






CD 






CJ 




cd 


E-h 






CD CJ 




CD 


3 






CD 


< 


CO 


CD 


Cn 




CD 


Eh 






CD 


< 


CO 


Q 


Eh 




CM 








CM E-l 


CO 


CN] 


CD 






CsJ 


CJ 




CM 


Cn CD 


CNl 


CD 






CN 


CJ 




CN] 


Eh 


ph ; 




cd 






y— < Eh 




Cs3 


E-h 


IS 




ro 


E-h 




^J 1 


-i-> 




LO 




CO 




LO 


CD 




C^- 


CD 






cj 


PS 




CJ 






< 








CJ 


1 — -1 




Cn 














CD 


O 




CJ 






cd 






CD 


< 




CD 








CD 






Cn CD 




Eh 








Eh 






< 


Eh ! 










CD 






CD 


CD 






Eh 






< 






CD 


> 






^5 






CJ 






£2 


s 




CD 






Eh 








E-h 


1 1 




CJ 






Eh 








<C 


CO 




CJ 


Oh ! 




cj 






CD 


CD 




E-h 








Eh 






Eh 


CO 




CD 








CJ 






CJ 




CJ 






E-h 






< 


i— 1 












CJ 








CO 






Eh 






< 






ro 






CJ 






CD 










1 — 1 




CJ 














Eh 


Cu 




o 




o 


o 






CD CJ) 




CD 


CD 






CD 


o 




CD 


Eh 


CO 


CD 


CJ 






CD 


CJ 




C3 


CJ 


Dh ; 


< — I 


o 






i — 1 CD 




i— 1 


Eh 






. \ — ( 






i— 1 


CJ 




i— 1 


Eh 


CO 




i— 1 


CD 












-t-j 


















ro 


Eh 


>H 


^H 


Eh 




LO 


CJ 






LO 


CJ 












+-> 












% 








CJ 






S3 


> 




CD 








Eh 






Eh 






ro 






CD 






CD 








CJ 












< 


CO 






CJ 






Eh 






ro 






Eh 


> 




Eh 








CD 


< 




CJ 






CJ 








<c 


E-i 










cn 






CD 




Eh 








< 






< 


Eh 




CJ 








Eh 






a 


CO : 




Eh 






CD 






CJ 


l-H 






CJ 






CJ 






CD 


< 






CJ 






CD 






CJ 






Eh 






<C 








<; 


Eh 




Eh 






Eh 






CJ 


D-, 




CJ 






CJ 






CJ 


t-H 




CD 








CJ 






CJ 






CD 








CJ 






< 


Eh ■ 



48 



EP 1 757 686 A1 



FIG. 10A 

LEADER SEQUENCE 



FIG. 10B 




FIG. 10C LYSATES CULTURE 

__ SLJPERNATANTS 

CD CO 

g Q § Q 

i — i — r*- 

uj O uu Q 

> CM > CM 



46 — 
30 ra 




49 



EP 1 757 686 A1 



FIG.11A 



Lf) 

CD 
CO 



o 

CM 



<0' :[■ 


" ■*? 


CM 


^sllllili 



r cd 



13> 

o 



CD CD 

*~ PI Log 



E 



ea 

o 

CO 



</3 
CD 
I 



CD 



•-..C4. ■ v.-.' . 

csi . . . . • 

o =.■•■■ - . :-. 


.0 >- .*** = 


o 


0°5 ■ 


ni'i ■ t f ^nnn i i ^mni i i 71m 1 1 1 1 

r*j •«— 0 
r> O CD CD 

^ PI Log "~ 


' ro-/"" ' ' -' : .' ; -\' ' ■ 
cni ^ • .' 
0 ■ ..• ■■• •. ■■ 


o-- = 






mrrmf — |imi i i 1 — "jm 1 1 1 i 


■ (iui 1 1 1 r 



it ^ o 



TO 
O 



- O 



o 

PI Log 



CM 

o 



rO 




rmn 1 " 1 pirm 1 ; — prrrnrt — pnrrnr 

■r> cm -— ri 

CD O CD CD 



O 
I 

O 



PI Log 







inn 1 1 1 


ITC Log 


0 tD -- 


•os '".•• = 

■T ■ ■. ; 










O 00 


ooo • • ■ 



CD 

PI Log 



pmT 1 "i " 

CD 



CD 
O 

O 















• ? - - 


'•0 -j 










-A. 
























nun 1 f 


• [mi ■ 


rn — pni III 


( — pmiT 1 t— 


0 


CM 

CD 




CD 






PI Log 





CJ5 
O 

O 



o 
o 



o 
o 



50 



EP 1 757 686 A1 



FIG. 11B 




51 



EP 1 757 686 A1 



FIG. 12A 



_ o 



oo 

CD 
CD 



< . . 


*«*-' : 


< in •■ , \.. 

pii ■ I* pfiitfr** — yiirrrf-r 

CD CD CD 

PI Log 


CD 






A1 
2.4 

■ 




mi hi i — jn-TrrT-« pmi i i i ■■ |iniiTTr 

CM * — <"> 



05 

o 
I 

O 



CD 



CD 
O 

o 



P! Log 



O ' : 

Osl ' 


■ 







CO 

CD 



CD 



o 



- CD 



o 



o 

PI Log 



<o. 



CO 

< in 



CO 



irntr-r" 
CD 



itt * p mn i 

CD 

PI Log 



_CD 



<o 



T" 

<_> 

CD 




o 

.CD — ' 



C 

o 
o 



< 

o 
o 



52 



EP 1 757 686 A1 



FIG. 12B 



i — 

I — 

«< 

I — 

o_ 

CO 



GO 

o 

CD 



<I . '.< -"v.-- •• ; 



T-OO-.-f ' 

< f ' , •■ 

— — n p un i i ~f — p un i i r 



if*- 




C33 
O 

O 



PI Log 



0>'. >>■.'•• 



A**"--" 



<j»*.: 



imiiTT i — [i n 1 1 1 i i — firm i t 




o 
o 



CO 

CD 



PI Log 



i — 

cn 

LU 
Q_ 

CO 

LU 

ce: 



co 

CD 




jii 1 1 it I t y nTTn 1 



PI Log 



CM 
< 



<00 



CM 

CD 



CD 



PI Log 



-c- O 



CD 

o 

CD — 1 



<Ccd ■ 




+ 



O 

o 



o 
o 



53 



EP 1 757 686 A1 



FIG. 13A 

VECTOR 2D7 DB 




ANNEX IN V ANNEX IN V 



FIG. 13B 



VECTOR 



i(r- 

10 2 - 
10 1 - 



A1 
6.8 




A2 
13.1 



A4>- 
8.0 



2D7 DB 



i ) i . i mii| i i uni 



10° 1"0 1 10 2 
ANNEX IN V 



10 J "|A1 

i 16.4 . 

10 2 i%V ; 4:.v; 




v", l '-r*4f*»r.- " 



10 d 



i i ■ ■ mi) I i i mi i | — i i rrrwq — i » imhi 

10° 10 1 10 2 10 
ANNEX IN V 



54 



EP 1 757 686 A1 



FIG. 13C 

VECTOR 



1 6.8 

10 2 



10 1 i 



10°^A3S. 
159 ' 




A2 
23 3 



5 Q.6y: ■■ . 



2D7 DB 



10*1aT— 

3l3.5 . 

io 2 i;:.^:.. ; - 

= 18.61 



i i'.iiiii I < i I I I. mi 

10 1 10 2 10 3 
ANNEX IN V 



A2 
50.5 




mi) 



MPrrr 



10° 10 1 10 2 
ANNEX IN V 



10 3 



FIG. 13D 

VECTOR 2D7 DB 




ANNEX IN V ANNEX IN V 



55 



EP 1 757 686 A1 




EP 1 757 686 A1 



FIG. 15A 



FIG. 15B 



£ 

=3 

o 



t£> - ! . . : . - 


CO 

<° 




l»r.#- .. 



o 

I — 

O 



C3 

PI Log 



o 



<C^^ :; ..-,: ; -..:-^« 



mi> 1 1 ( — ptrrrm — [mm ■ r pTTT. . i v 



_ CD 



■*t-'.-..„ 




cn 
o 
i 

O 



PI Log 



3 

in 
o 





O 




ill 



PI Log 



o 

t — 

O 



O 





r 
> 


fx»i i * rfiri i i — wjii t" "i 







co 
o 

C3 — 1 



PI Log 



E 



-<3- . - -\ 


r 

<o 







PI Log 



CD 

o 

■_J 

O 



£ 
o 

O 



u.1.11 1 > prrm r f »n 



5&. 



PI Log 




co 
o 

,-J 
O 



57 



EP 1 757 686 A1 



FIG. 15C FIG. 15D 




CD CD CD CD C5 CD CD CD 



PI Log PI Log 



58 



EP 1 757 686 A1 



FIG. 16A 



0 ug/ml 




"i i mi ] i' I i l ii i ij — i i i rrnt] — i )"i r ii u 

10° 10 1 10 2 10 : 
FITC Log 



10^ 



2.0 ug/ml 



|A1 ■ 

133.0% 



O 



A2 : 
40.4% 




FITC Log 



FIG. 16B 



10' 



§A1 

=11.9% 



0 ug/ml 

A2 

22.6% ■ ;-. •_ • 





I 'I "f I ft III — 1 I I mill — TTT 



FITC Log 



Trnt] — i i mm. 
10 2 10 



10 1ST 



2.0 ug/ml 



o 




1.0 1 - 



38.8% g^; '[47% 



r 1 1 iiiiii — TTT 



FITC Log 



59 



EP 1 757 686 A1 



FIG. 17A 



10%; 



A1 
1 7.5% 



10- 



o 



10 1 i 



0 HOUR 



>A2 
3.0% 




A4 
0.1% 



12 HOURS 



( nuii| — i i i rraij — i i i mil 

1 10 2 



10 u 10 
FITC Log 



10° 




FITC Log 



24 HOURS 



10^ 



iai m 

66.4% -:J2.1% ' 




38 HOURS 




i ■ mill — ~i T 



— i i mm) — i 1 1 iimj 

10 1 10 2 10 3 
FITC Log 




FITC Log 



60 



EP 1 757 686 A1 



FIG. 17B 



0 HOUR 



12 HOURS 




10 J 



-43.5% 



10%t 



O 



10 



1_ 



io°4 



A2 ■ 
1.7% 




5A3J 

:54 8% r 



ST 



A4 
0.0% 



i i t tnin i ) i i i i i iuj — i i mm 



FITC Log 



10 u 10 1 10 2 
FITC Log 



10° 



24 HOURS 



38 HOURS 




FITC Log 



10 u 10' 
FITC Log 



61 



EP 1 757 686 A1 




62 



EP 1 757 686 A1 



FIG. 19 



E 

"B> ■ 

>- 

CD 

+ 



o 
to 

u_ 
a 

+ 



CO CM , 



rnn i i~i — jnrm i r 



PI Log:: 




>- 

CD 



C-5 



<N ■«- 
CD W> . ■ 



Cvl 
^- CO 



111! 1 1 I I mi i M t r 




jfll Hit 

3 ■ 



E 
"ai 

ZD 

CsJ, 

CD 
t*- 

Q 

CM 
+ 




CO 



;i m i i i —i — pn I i i — [mm i 



-5b 



I [Ills 

IS 



CD 

CO 

m ■«— 



rn-T 



P I Log 



PI Log 



-a- • 
cn< • ; 
m ■■- •■ 



■:■■;:.<: ..- 



CO 



□2: 



imiTn — |in 1 1 1 i i — prm r i — 'jimr 

C3 C> 'o 




3b 



PI Log 



63 



EP 1 757 686 A1 



FIG. 20 



?1 

O 

EE ^ 



5 Q 



> 

i 

ni 
+ 



C_5 . , 















V 






■' 








t« is , • . 

' . •" > 




B1 

20.6% 







~1 p it i I T I 

Pirog 




E 




PI Log 




PI Log 



PI Log 




64 



EP 1 757 686 A1 



FIG. 21 



ARH77 



Jurkat 























CD 


CD 


LU 


CD 


>- 












CD 


LU 

















































CD 


CD 


LU 


CO 


>- 












CZ3 


l_U 






or 








I — 


CD 











65 



EP 1 757 686 A1 



FIG. 22 



80 




0 ' ' ' ' ' — 

0 200 500 1000 



2D7DB ( ng/ml ) 



66 



EP 1 757 686 A1 




67 



EP 1 757 686 A1 




68 



EP 1 757 686 A1 



FIG. 25 



100 



80 



S= 60 



40 
20 



0 



0 



20 40 60 80 100 120 

DAYS AFTER TUMOR TRANSPLANTATION 



140 



VEH I CLE 



•2D7 DIABODY 



69 



EP 1 757 686 A1 



FIG. 26 A 



FIG. 26B 



_o 



























\..^7£.'-. ' 





File: 

20021 195.LMD 
Quad % Total 



1(T 



10 10 
FITC 



10 10 



UL 
UR 
LL 
LR 



4.94 
20.26 
65.33 

9.47 



E^ 



■ mi'ii 

io 6 To r 



1 

10 
FITC 



i • ■•■■■i.vS >/ 



File: 

20021 198.LMD 
Quad % Total 



2 10 3 10 4 



UL 
UR 
LL 
LR 



2.04 
22.04 
65.98 

9.94 




File: 

20021 194.LMD 
Quad % Total 



10' 10' 
FITC 



UL 
UR 
LL 
LR 



7.23 
45.44 
36.01 
11.32 



— o 



10 



10 1 10 2 
FITC 



trwr 



10 J 



File: 

200211 97. LMD 
Quad % Total 



To 4 



UL 
UR 
LL 
LR 



2.37 
30.29 
52.65 
14.69 



■ 

■ * 

i - • 


. V- 
#\ 


: wga 

1 





10' 



File: 

20021 193.LMD 


■ 

CO • 

o 


Quad % Total 


_ — 1 
■ 


UL 6.89 
UR 42.35 


■ 

t— - 


LL 40.76 


pi 

: 

o : 
o_ 


LR 10.00 



FITC 



10° 



10 u 



File: 

20021 1 96.LMD 
Quad % Total 



FITC 



10 10 



UL 
UR 
LL 
LR 



3.16 
33.60 
48.20 
15.04 



70 



EP 1 757 686 A1 



FIG. 26C 



FIG. 26D 




File: 

Z0021202.LMD 



Quad 


% Total 


UL 


2.07 


UR 


31.24 


LL 


56.25 


LR 


10.44 



10 10 10" 



FITC 



5^ 




10" 



T»rn i m if-fiiia 

10 1 10 2 10 3 
FITC 



File: 

Z0021205.LMD 



Quad 


% Total 


UL 


1.20 


UR 


28.74 


LL 


63.74 


LR 


6.32 



10^ 




File: 

Z0021201.LMD 



Quad 


% Total 


UL 


1.98 


UR 


37.69 


LL 


30.44 


LR 


29.89 



10 10 
FITC 



_o. 



3 .v??w*v.. 



■mi* 



10" 



10' 10 2 
FITC 



File: 

Z0021204.LMD 



Quad 


% Total 


UL 


0.91 


UR 


22.71 


LL 


40.28 


LR 


36.10 



10 10 



.... . 



ftf I H l l | 



10" 



10 10 
FITC 



10' 



File: 

Z0021199.LMD 
Quad % Total 



UL 
UR 
LL 
LR 



10' 



2.46 
40.37 
31.00 
26.17 



_o. 









ft>y?'r:' — 
- , 



10 u 10 1 



File: 

Z0021203.LMD 



Quad 


% Total 


UL 


0.93 


UR 


24.60 


LL 


42.03 


LR 


32.44 



FITC 



71 



EP 1 757 686 A1 



FIG. 26E 



o 



_o 



o 
O. 



■ 

■ 
■ 


a 

. •* 
- j _ ■ • * 


File: Z0021208.LMD 

Quad % Total 
UL 1.95 






UR 


15.84 






LL 


77.05 






LR 


5.16 



10° 10 1 10 2 10 3 10 4 
FITC 



o 



CO 

o 



CM 

o 



o 
O. 



■ 


• - . 


* ., • 


• 



File: 20021 207. LMD 



Quad 


% Total 


UL 


1.51 


UR 


55.82 


LL 


27.05 


LR 


15.62 



10 u 



10 10 
FITC 



10 10 



o 



CM 

o 



o 



' . " 


» .5 


S » * . • « I | • 
1 • • . m 





File: Z0021206.LMD 
Quad % Total 



10 L 



10 10 
FITC 



10 3 10 4 



UL 
UR 
LL 
LR 



1.40 
57.54 
20.64 
20.42 



72 



EP 1 757 686 A1 



INTERN ATIONAL SEARCH REPORT 



International application No. 

PCT/JP2004/005152 



A. CLASSIFICATION OF SUBJECT MATTER 

Int. CI 7 C12N15/09, C07K16/28, C07K16/46, A61P35/00, A61P37/02, 
A61P43/00, A61K39/395 

According to International Patent Classification (IPC) or to both national classification and IPC 



B. FIELDS SEARCHED 



Minimum documentation searched (classification system followed by classification symbols) 

Int. CI 7 C12N15/09, C07K16/28, C07K16/46, A61P35/00, A61P37/02, 
A61P43/00, A61K39/395 



Documentation searched other than minimum documentation to the extent that such documents are included in the fields searched 



Electronic data base consulted during the international search (name of data base and, where practicable, search terms used) - 

MEDLINE, BIOSIS/WPI (DIALOG) , SwissProt/PIR/GeneSeq, Genbank/EMBL/DDB J 



C. DOCUMENTS CONSIDERED TO BE RELEVANT 



Category* 


Citation of document, with indication, where appropriate, of the relevant passages 


Relevant to claim No. 


Y 


Hudson P.J. et al., High avidity scFv multimers; 
diabodies and triabodies, J . Immunol .Methods, 
1999, Vol.231, pages 177 to 189 


1- 


14 


Y 


Kortt A. A. et al . , Dimeric and trimeric anti 
bodies: high avidity scFvs for cancer target 
ing, Biomol.Eng., 2001, Vol.18, pages 95 to 
10 8 


1- 


14 


Y 


Xiong D. et al., Efficient inhibition of human 
B-cell lymphoma xenografts with an anti-CD2 0 x 
anti-CD3 bispecific diabody, Cancer Lett., 2002, 
Vol.177, pages 29 to 39 


1- 


14 



I x I Further documents are listed in the continuation of Box C. 



I I See patent family annex. 



"A" 



"O" 
"P" 



Special categories of cited documents: "T f 
document defining the general state of the art which is not considered 
to be of particular relevance 

earlier application or patent but published on or after the international ' "X" 
filing date 

document which may throw doubts on priority claim(s) or which is 

cited to establish the publication date of another citation or other «-y» 

special reason (as specified) 

document referring to an oral disclosure, use, exhibition or other means 
document published prior to the international filing date but later than 
the priority date claimed 



later document published after the international filing date or priority 
date and not in conflict with the application but cited to understand 
the principle or theory underlying the invention 

document of particular relevance; the claimed invention cannot be 
considered novel or cannot be considered to involve an inventive 
step when the document is taken alone 

document of particular relevance; the claimed invention cannot be 
considered to involve an inventive step when the document is 
combined with one or more other such documents, such combination 
being obvious to a person skilled in the art 
document member of the same patent family 



Date of the actual completion of the international search 
05 July, 2004 (05.07.04) 


Date of mailing of the international search report 
20 July, 2004 (20.07.04) 


Name and mailing address of the ISA/ 

Japanese Patent Office 

Facsimile No. 


Authorized officer 
Telephone No. 



Form PCT/ISA/210 (second sheet) (January 2004) 



73 



EP 1 757 686 A1 



INTERNATIONAL SEARCH REPORT 



International application No. 

PCT/JP2004/005152 



C (Continuation). DOCUMENTS CONSIDERED TO BE RELEVANT 



Category* 



E,X 



Citation of document, with indication, where appropriate, of the relevant passages 



MATSUOKA S. et al . , A novel type of cell death 
of lymphocytes induced, by a monoclonal antibody 
without participation of complement, J. Exp. Med., 
1995, Vol.181, pages 2007 to 2015 

Fayen J. et al.. Negative signaling by anti- 
HLA claas I antibodies is dependent upon two 
triggering events, Int . Immunol . , 1998, Vol.10, 
pages 1347 to 1358 

Woodle E.S. et al.. Anti-human class I MHC 
antibodies induce apoptosis by a pathway that 
is distinct from the Fas antigen-mediated path 
way, J. Immunol., 1997, Vol.158, pages 2156 to 
2164 

Tahtis k. et al . , Biodistribution properties 
of (111) indium-labeled C-functionalized 
trans -cyclohexyl diethylenetriaminepentaacetic 
acid humanized 3S193 diabody and F(ab') (2), 
constructs in a breast carcinoma xenograft 
model, Clin. Cancer Res., 20 01, Vol.7, pages 
1061 to 1072 

Rossi E.A. et al . , Development of new multi 
valent-bispecif ic agents for pretargeting 
tumor localization and therapy, Clin. Cancer 
Res., 2003, Vol.9, pages 38865 to 3896S 

WO 04/0334 99 Al (Chugai Pharmaceutical Co., 
Ltd. ) , 

22 April, 2004 (22.04.04), 
Full text 
(Family: none) 



Relevant to claim No. 



1-14 



1-14 



1-14 



1-U 



1-14 



1-14 



Form PCT/ISA/210 (continuation of second sheet) (January 2004) 



74 



EP 1 757 686 A1 



REFERENCES CITED IN THE DESCRIPTION 

This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European 
patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be 
excluded and the EPO disclaims all liability in this regard. 



Patent documents cited in the description 

EP 404097 A [0019] 
WO 9311161 A [0019] 
WO 9846777 A [0030] 
EP 239400 A [0032] 
WO 9602576 A [0032] 
JP 1059878 B [0034] 
WO 9312227 A [0034] 
WO 9203918 A [0034] 
WO 9402602 A [0034] 
WO 9425585 A [0034] 
WO 9634096 A [0034] 



WO 9633735 A [0034] 
WO 9201 047 A [0034] 
WO 9220791 A [0034] 
WO 9306213 A [0034] 
WO 9311236 A [0034] 
WO 9319172 A [0034] 
WO 9501438 A [0034] 
WO 9515388 A [0034] 
US 5057313 A [0062] 
US 5156840 A [0062] 



Non-patent literature cited in the description 

FAYEN et al. Int. Immunol., 1 998, vol. 1 0, 1 347-1 358 
[0007] 

GENESTIER et al. Blood, 1997, vol. 90, 3629-3639 
[0007] 

GENESTIER et al. Blood, 1997, vol. 90, 726-735 
[0007] 

GENESTIER et al. J. Biol. Chem., 1998, vol. 273, 
5060-5066 [0007] 

WOODLE et al. J. Immunol., 1997, vol. 158, 
2156-2164 [0007] 

MATSUOKA et al. J. Exp. Med., 1995, vol. 181, 
2007-2015 [0007] 

GOTO et al. Blood, 1 994, vol. 84, 1 922 [0007] 
HUSTON, J. S. et al. Proc. Natl. Acad. Sci. U.S.A., 

1988, vol. 85, 5879-5883 [0016] 
PLICKTHUN. The Pharmacology of Monoclonal An- 
tibodies. Springer Verlag, 1994, vol. 113, 269-315 
[0016] 

CO, M. S. et al. J. Immunol., 1994, vol. 152, 
2968-2976 [0016] 

• BETTER, M. ; HORWITZ, A. H. Methods Enzymol., 

1989, vol. 178, 476-496 [0016] 
PLUCKTHUN, A. ; SKERRA, A. Methods Enzymol., 
1989, vol. 178, 497-515 [0016] 

LAMOYI, E. Methods Enzymol., 1986, vol. 121, 
652-663 [0016] 

ROUSSEAUX, J. et al. Methods Enzymol., 1 986, 
vol. 121, 663-669 [0016] 

• BIRD, R. E. ; WALKER, B. W. Trends Biotechnol., 
1991, vol. 9, 132-137 [0016] 

• P. HOLLIGER et al. Proc. Natl. Acad. Sci. USA, 
1993, vol. 90, 6444-6448 [0019] 

JOHNSON et al. Method in Enzymology, 1 991 , vol. 
203, 88-98 [0019] 



HOLLIGER et al. Protein Engineering, 1 996, vol. 9, 
299-305 [0019] 

PERISIC et al. Structure, 1994, vol. 2, 1217-1226 
[0019] 

JOHN et al. Protein Engineering, 1999, vol. 12 (7), 
597-604 [0019] 

HOLLIGER et al. Proc. Natl. Acad. Sci. USA., 1 993, 
vol. 90, 6444-6448 [0019] 

ATWELL et al. Mol. Immunol., 1996, vol. 33, 
1301-1312 [0019] 

MARK, D. F. et al. Proc. Natl. Acad. Sci. USA, 1 984, 
vol. 81 , 5662-5666 [0028] 

ZOLLER, M. J. ; SMITH, M. Nucleic Acids Research, 
1982, vol. 10, 6487-6500 [0028] 
WANG, A. et al. Science, vol. 224, 1 431 -1 433 [0028] 
DALBADIE-MCFARLAND, G. et al. Proc. Natl. 
Acad. Sci. USA, 1982, vol. 79, 6409-6413 [0028] 
• KOHLER, G. ; MILSTEIN, C. Methods Enzymol., 
1981, vol. 73, 3-46 [0030] 

SATO, K. et al. Cancer Res., 1 993, vol. 53, 851 -856 
[0032] 

SAMBROOK, J. et al. Molecular Cloning. Cold 
Spring Harbor Lab. press, 1989, 9.47-9.58 [0037] 
WARD et al. Nature, 1 989, vol. 341 , 544-546 [0041 ] 
FASEB J., 1 992, vol. 6, 2422-2427 [0041] 
BETTER et al. Science, 1 988, vol. 240, 1 041 -1 043 
[0041] 

LEI, S. P. et al. J. Bacteriol., 1987, vol. 169, 4379 
[0042] 

Nucleic Acids Res., 1 990, vol. 1 8 (1 7), 5322 [0043] 
MULLIGAN et al. Nature, 1 979, vol. 277, 1 08 [0044] 
MIZUSHIMA et al. Nucleic Acids Res., 1 990, vol. 1 8, 
5322 [0044] 

J. Exp. Med., 1 995, vol. 1 08, 945 [0048] 



75 



EP 1 757 686 A1 



VALLE et al. Nature, 1 981 , vol. 291 , 358-340 [0048] 
Proc. Natl. Acad. Sci. USA, 1 980, vol. 77, 421 6-4220 
[0048] 

Proc. Natl. Acad. Sci. USA, 1968, vol. 60, 1275 
[0048] 

EBERT, K.M. et al. Bio/Technology, 1994, vol. 12, 
699-702 [0054] 

SUSUMU, M. et al. Nature, 1985, vol. 315, 592-594 
[0055] 



JULIAN K.-C. MA et al. Eur. J. Immunol., 1 994, vol. 
24, 131-138 [0056] 

Strategies for Protein Purification and Characteriza- 
tion: A Laboratory Course Manual. Cold Spring Har- 
bor Laboratory Press, 1 996 [0058] 
Antibodies A Laboratory Manual. Cold Spring Harbor 
Laboratory, 1 988 [0059] 



76